351
|
Wang SS, Zhang QL, Chu P, Kong LQ, Li GZ, Li YQ, Yang L, Zhao WJ, Guo XH, Tang ZY. Synthesis and antitumor activity of α,β-unsaturated carbonyl moiety- containing oleanolic acid derivatives targeting PI3K/AKT/mTOR signaling pathway. Bioorg Chem 2020; 101:104036. [DOI: 10.1016/j.bioorg.2020.104036] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 04/24/2020] [Accepted: 06/19/2020] [Indexed: 12/22/2022]
|
352
|
PROTACs: An Emerging Therapeutic Modality in Precision Medicine. Cell Chem Biol 2020; 27:998-1014. [DOI: 10.1016/j.chembiol.2020.07.020] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/17/2020] [Accepted: 07/23/2020] [Indexed: 12/17/2022]
|
353
|
KRAS(G12C)-AMG 510 interaction dynamics revealed by all-atom molecular dynamics simulations. Sci Rep 2020; 10:11992. [PMID: 32686745 PMCID: PMC7371895 DOI: 10.1038/s41598-020-68950-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/03/2020] [Indexed: 01/02/2023] Open
Abstract
The first KRAS(G12C) targeting inhibitor in clinical development, AMG 510, has shown promising antitumor activity in clinical trials. On the molecular level, however, the interaction dynamics of this covalently bound drug–protein complex has been undetermined. Here, we disclose the interaction dynamics of the KRAS(G12C)–AMG 510 complex by long timescale all-atom molecular dynamics (MD) simulations (total of 75 μs). Moreover, we investigated the influence of the recently reported post-translational modification (PTM) of KRAS’ N-terminus, removal of initiator methionine (iMet1) with acetylation of Thr2, to this complex. Our results demonstrate that AMG 510 does not entrap KRAS into a single conformation, as one would expect based on the crystal structure, but rather into an ensemble of conformations. AMG 510 binding is extremely stable regardless of highly dynamic interface of KRAS’ switches. Overall, KRAS(G12C)–AMG 510 complex partially mimic the native dynamics of GDP bound KRAS; however, AMG 510 stabilizes the α3-helix region. N-terminally modified KRAS displays similar interaction dynamics with AMG 510 as when Met1 is present, but this PTM appears to stabilize β2–β3-loop. These results provide novel conformational insights on the molecular level to KRAS(G12C)–AMG 510 interactions and dynamics, providing new perspectives to RAS related drug discovery.
Collapse
|
354
|
Avoiding or Co-Opting ATP Inhibition: Overview of Type III, IV, V, and VI Kinase Inhibitors. NEXT GENERATION KINASE INHIBITORS 2020. [PMCID: PMC7359047 DOI: 10.1007/978-3-030-48283-1_3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
As described in the previous chapter, most kinase inhibitors that have been developed for use in the clinic act by blocking ATP binding; however, there is growing interest in identifying compounds that target kinase activities and functions without interfering with the conserved features of the ATP-binding site. This chapter will highlight alternative approaches that exploit unique kinase structural features that are being targeted to identify more selective and potent inhibitors. The figure below, adapted from (Sammons et al., Molecular Carcinogenesis 58:1551–1570, 2019), provides a graphical description of the various approaches to manipulate kinase activity. In addition to the type I and II inhibitors, type III kinase inhibitors have been identified to target sites adjacent to the ATP-binding site in the catalytic domain. New information on kinase structure and substrate-binding sites has enabled the identification of type IV kinase inhibitor compounds that target regions outside the catalytic domain. The combination of targeting unique allosteric sites outside the catalytic domain with ATP-targeted compounds has yielded a number of novel bivalent type V kinase inhibitors. Finally, emerging interest in the development of irreversible compounds that form selective covalent interactions with key amino acids involved in kinase functions comprise the class of type VI kinase inhibitors.
Collapse
|
355
|
Ertl P, Altmann E, McKenna JM. The Most Common Functional Groups in Bioactive Molecules and How Their Popularity Has Evolved over Time. J Med Chem 2020; 63:8408-8418. [DOI: 10.1021/acs.jmedchem.0c00754] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Peter Ertl
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| | - Eva Altmann
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Basel CH-4056, Switzerland
| | - Jeffrey M. McKenna
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
356
|
Hassaan E, Hohn C, Ehrmann FR, Goetzke FW, Movsisyan L, Hüfner-Wulsdorf T, Sebastiani M, Härtsch A, Reuter K, Diederich F, Klebe G. Fragment Screening Hit Draws Attention to a Novel Transient Pocket Adjacent to the Recognition Site of the tRNA-Modifying Enzyme TGT. J Med Chem 2020; 63:6802-6820. [PMID: 32515955 DOI: 10.1021/acs.jmedchem.0c00115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Fragment-based lead discovery was applied to tRNA-guanine transglycosylase, an enzyme modifying post-transcriptionally tRNAs in Shigella, the causative agent of shigellosis. TGT inhibition prevents translation of Shigella's virulence factor VirF, hence reducing pathogenicity. One discovered fragment opens a transient subpocket in the preQ1-recognition site by pushing back an aspartate residue. This step is associated with reorganization of further amino acids structurally transforming a loop adjacent to the recognition site by duplicating the volume of the preQ1-recognition pocket. We synthesized 6-carboxamido-, 6-hydrazido-, and 4-guanidino-benzimidazoles to target the opened pocket, including a dihydro-imidazoquinazoline with a propyn-1-yl exit vector pointing into the transient pocket and displacing a conserved water network. MD simulations and hydration-site analysis suggest water displacement to contribute favorably to ligand binding. A cysteine residue, exclusively present in bacterial TGTs, serves as gatekeeper of the transient subpocket. It becomes accessible upon pocket opening for selective covalent attachment of electrophilic ligands in eubacterial TGTs.
Collapse
Affiliation(s)
- Engi Hassaan
- Institute of Pharmaceutical Chemistry, University of Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| | - Christoph Hohn
- Laboratorium für Organische Chemie, ETH Zurich, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland
| | - Frederik R Ehrmann
- Institute of Pharmaceutical Chemistry, University of Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| | - F Wieland Goetzke
- Laboratorium für Organische Chemie, ETH Zurich, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland
| | - Levon Movsisyan
- Laboratorium für Organische Chemie, ETH Zurich, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland
| | - Tobias Hüfner-Wulsdorf
- Institute of Pharmaceutical Chemistry, University of Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| | - Maurice Sebastiani
- Institute of Pharmaceutical Chemistry, University of Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| | - Adrian Härtsch
- Laboratorium für Organische Chemie, ETH Zurich, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland
| | - Klaus Reuter
- Institute of Pharmaceutical Chemistry, University of Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| | - François Diederich
- Laboratorium für Organische Chemie, ETH Zurich, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland
| | - Gerhard Klebe
- Institute of Pharmaceutical Chemistry, University of Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| |
Collapse
|
357
|
Chen H, Liu L, Huang T, Chen J, Chen T. Direct Dehydrogenation for the Synthesis of α,β‐Unsaturated Carbonyl Compounds. Adv Synth Catal 2020. [DOI: 10.1002/adsc.202000454] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Hong Chen
- Haikou Hospital affiliated to Xiangya School of MedicineCentral South University Haikou 570100 People's Republic of China
| | - Long Liu
- Key Laboratory of Ministry of Education for Advanced Materials in Tropical Island ResourcesHainan Provincial Key Lab of Fine ChemHainan University Haikou 570228 People's Republic of China
| | - Tianzeng Huang
- Key Laboratory of Ministry of Education for Advanced Materials in Tropical Island ResourcesHainan Provincial Key Lab of Fine ChemHainan University Haikou 570228 People's Republic of China
| | - Jing Chen
- Haikou Hospital affiliated to Xiangya School of MedicineCentral South University Haikou 570100 People's Republic of China
| | - Tieqiao Chen
- Key Laboratory of Ministry of Education for Advanced Materials in Tropical Island ResourcesHainan Provincial Key Lab of Fine ChemHainan University Haikou 570228 People's Republic of China
| |
Collapse
|
358
|
Zhang X, Fang W, Lekkala R, Tang W, Qin H. An Easy, General and Practical Method for the Construction of Alkyl Sulfonyl Fluorides. Adv Synth Catal 2020. [DOI: 10.1002/adsc.202000515] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Xu Zhang
- State Key Laboratory of Silicate Materials for Architecturesand School of ChemistryChemical Engineering and Life ScienceWuhan University of Technology 205 Luoshi Road Wuhan 430070 People's Republic of China
| | - Wan‐Yin Fang
- State Key Laboratory of Silicate Materials for Architecturesand School of ChemistryChemical Engineering and Life ScienceWuhan University of Technology 205 Luoshi Road Wuhan 430070 People's Republic of China
| | - Ravindar Lekkala
- State Key Laboratory of Silicate Materials for Architecturesand School of ChemistryChemical Engineering and Life ScienceWuhan University of Technology 205 Luoshi Road Wuhan 430070 People's Republic of China
| | - Wenjian Tang
- School of PharmacyAnhui Province Key Laboratory of Major Autoimmune DiseasesAnhui Medical University Hefei 230032 People's Republic of China
| | - Hua‐Li Qin
- State Key Laboratory of Silicate Materials for Architecturesand School of ChemistryChemical Engineering and Life ScienceWuhan University of Technology 205 Luoshi Road Wuhan 430070 People's Republic of China
| |
Collapse
|
359
|
Sutanto F, Konstantinidou M, Dömling A. Covalent inhibitors: a rational approach to drug discovery. RSC Med Chem 2020; 11:876-884. [PMID: 33479682 PMCID: PMC7557570 DOI: 10.1039/d0md00154f] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 06/19/2020] [Indexed: 12/13/2022] Open
Abstract
In this review we provide a brief historic overview of covalent inhibitors and summarize recent advances focusing on developments in the last decade. Applications in challenging targets and future perspectives are also discussed.
Covalent inhibitors are recognized as an important component in drug discovery and therapeutics. Since the first appearance of covalent inhibitors in the late 18th century, the field has advanced significantly and currently about 30% of the marketed drugs are covalent inhibitors. The numerous advantages of covalent inhibitors are counteracting the initial concerns regarding potential off-target toxicity. Thus, continuous research, especially for cancer targets is reported. The aim of this review is to provide a short historic overview and focus on recently developed covalent inhibitors (2011–2019), including structural aspects and examples on challenging targets.
Collapse
Affiliation(s)
- Fandi Sutanto
- Department of Pharmacy , Group of Drug Design , University of Groningen , A. Deusinglaan 1 , 9713 AV , Groningen , The Netherlands . ; http://www.drugdesign.nl
| | - Markella Konstantinidou
- Department of Pharmacy , Group of Drug Design , University of Groningen , A. Deusinglaan 1 , 9713 AV , Groningen , The Netherlands . ; http://www.drugdesign.nl
| | - Alexander Dömling
- Department of Pharmacy , Group of Drug Design , University of Groningen , A. Deusinglaan 1 , 9713 AV , Groningen , The Netherlands . ; http://www.drugdesign.nl
| |
Collapse
|
360
|
Mulder T, Bobba S, Johnson K, Grandner JM, Wang W, Zhang C, Cai J, Choo EF, Khojasteh SC, Zhang D. Bioactivation of α,β-Unsaturated Carboxylic Acids Through Acyl Glucuronidation. Drug Metab Dispos 2020; 48:819-829. [DOI: 10.1124/dmd.120.000096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/23/2020] [Indexed: 01/06/2023] Open
|
361
|
Al-Awadhi FH, Luesch H. Targeting eukaryotic proteases for natural products-based drug development. Nat Prod Rep 2020; 37:827-860. [PMID: 32519686 PMCID: PMC7406119 DOI: 10.1039/c9np00060g] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Covering: up to April 2020 Proteases are involved in the regulation of many physiological processes. Their overexpression and dysregulated activity are linked to diseases such as hypertension, diabetes, viral infections, blood clotting disorders, respiratory diseases, and cancer. Therefore, they represent an important class of therapeutic targets. Several protease inhibitors have reached the market and >60% of them are directly related to natural products, even when excluding synthetic natural product mimics. Historically, natural products have been a valuable and validated source of therapeutic agents, as over half of the marketed drugs across targets and diseases are inspired by natural product structures. In the past two decades the number of new protease inhibitors discovered from nature has sharply increased. Additionally, the availability of 3D structural information for proteases has permitted structure-based design and accelerated the synthesis of optimized lead structures with improved potency and selectivity profiles, resulting in some of the most-potent-in-class inhibitors. These discoveries were oftentimes maximized by in-depth biological assessments of lead inhibitors, linking them to a relevant disease state. This review will discuss some of the current and emerging drug targets and their involvement in various disease processes, highlighting selected success stories behind several FDA-approved protease inhibitors that have natural products scaffolds as well as recent selected pharmacologically well-characterized inhibitors derived from marine or terrestrial sources.
Collapse
Affiliation(s)
- Fatma H Al-Awadhi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait.
| | - Hendrik Luesch
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, 1345 Center Drive, Gainesville, Florida 32610, USA.
| |
Collapse
|
362
|
Abstract
RAS (KRAS, NRAS and HRAS) is the most frequently mutated gene family in cancers, and, consequently, investigators have sought an effective RAS inhibitor for more than three decades. Even 10 years ago, RAS inhibitors were so elusive that RAS was termed 'undruggable'. Now, with the success of allele-specific covalent inhibitors against the most frequently mutated version of RAS in non-small-cell lung cancer, KRASG12C, we have the opportunity to evaluate the best therapeutic strategies to treat RAS-driven cancers. Mutation-specific biochemical properties, as well as the tissue of origin, are likely to affect the effectiveness of such treatments. Currently, direct inhibition of mutant RAS through allele-specific inhibitors provides the best therapeutic approach. Therapies that target RAS-activating pathways or RAS effector pathways could be combined with these direct RAS inhibitors, immune checkpoint inhibitors or T cell-targeting approaches to treat RAS-mutant tumours. Here we review recent advances in therapies that target mutant RAS proteins and discuss the future challenges of these therapies, including combination strategies.
Collapse
|
363
|
Leng J, Tang W, Fang WY, Zhao C, Qin HL. A Simple Protocol for the Stereoselective Construction of Enaminyl Sulfonyl Fluorides. Org Lett 2020; 22:4316-4321. [PMID: 32407099 DOI: 10.1021/acs.orglett.0c01360] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
A clickable connective hub 1-bromo-2-triazolethane-1-sulfonyl fluoride BTESF (1) was developed and successfully applied for the fluorosulfonylvinylation of a host of primary and secondary cyclic or acyclic amines including amino acids and pharmaceuticals. Further antimicrobial experiments revealed that vinyl sulfonyl fluoride functionalized norfloxacin (3ak), ciprofloxacin (3am), and lomefloxacin (3an) exhibited 4-fold improved antimicrobial activity against Gram-positive bacteria compared to their parent drugs.
Collapse
Affiliation(s)
- Jing Leng
- State Key Laboratory of Silicate Materials for Architectures; and School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, 205 Luoshi Road, Wuhan 430070, P.R. China
| | - Wenjian Tang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Wan-Yin Fang
- State Key Laboratory of Silicate Materials for Architectures; and School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, 205 Luoshi Road, Wuhan 430070, P.R. China
| | - Chuang Zhao
- State Key Laboratory of Silicate Materials for Architectures; and School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, 205 Luoshi Road, Wuhan 430070, P.R. China
| | - Hua-Li Qin
- State Key Laboratory of Silicate Materials for Architectures; and School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, 205 Luoshi Road, Wuhan 430070, P.R. China
| |
Collapse
|
364
|
McAulay K, Hoyt EA, Thomas M, Schimpl M, Bodnarchuk MS, Lewis HJ, Barratt D, Bhavsar D, Robinson DM, Deery MJ, Ogg DJ, Bernardes GJL, Ward RA, Waring MJ, Kettle JG. Alkynyl Benzoxazines and Dihydroquinazolines as Cysteine Targeting Covalent Warheads and Their Application in Identification of Selective Irreversible Kinase Inhibitors. J Am Chem Soc 2020; 142:10358-10372. [PMID: 32412754 DOI: 10.1021/jacs.9b13391] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
With a resurgence in interest in covalent drugs, there is a need to identify new moieties capable of cysteine bond formation that are differentiated from commonly employed systems such as acrylamide. Herein, we report on the discovery of new alkynyl benzoxazine and dihydroquinazoline moieties capable of covalent reaction with cysteine. Their utility as alternative electrophilic warheads for chemical biological probes and drug molecules is demonstrated through site-selective protein modification and incorporation into kinase drug scaffolds. A potent covalent inhibitor of JAK3 kinase was identified with superior selectivity across the kinome and improvements in in vitro pharmacokinetic profile relative to the related acrylamide-based inhibitor. In addition, the use of a novel heterocycle as a cysteine reactive warhead is employed to target Cys788 in c-KIT, where acrylamide has previously failed to form covalent interactions. These new reactive and selective heterocyclic warheads supplement the current repertoire for cysteine covalent modification while avoiding some of the limitations generally associated with established moieties.
Collapse
Affiliation(s)
| | - Emily A Hoyt
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
| | | | - Marianne Schimpl
- Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge CB4 0WG, U.K
| | | | | | - Derek Barratt
- Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Deepa Bhavsar
- Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | | | - Michael J Deery
- Cambridge Centre for Proteomics, Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, U.K
| | - Derek J Ogg
- Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Gonçalo J L Bernardes
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.,Instituto de Medicina Molecular, Faculdade de Medicina de Universidad de Lisboa, Avenida Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | | | - Michael J Waring
- Northern Institute for Cancer Research, Chemistry, School of Natural and Environmental Sciences, Newcastle University, Bedson Building, Newcastle upon Tyne NE1 7RU, U.K
| | | |
Collapse
|
365
|
|
366
|
Huang F, Hu H, Wang K, Peng C, Xu W, Zhang Y, Gao J, Liu Y, Zhou H, Huang R, Li M, Shen J, Xu Y. Identification of Highly Selective Lipoprotein-Associated Phospholipase A2 (Lp-PLA2) Inhibitors by a Covalent Fragment-Based Approach. J Med Chem 2020; 63:7052-7065. [DOI: 10.1021/acs.jmedchem.0c00372] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Fubao Huang
- State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hangchen Hu
- CAS Key Laboratory of Receptor Research, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kai Wang
- State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Chengyuan Peng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wenwei Xu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yu Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jing Gao
- CAS Key Laboratory of Receptor Research, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yishen Liu
- Nanchang University, Nanchang 330031, China
| | - Hu Zhou
- CAS Key Laboratory of Receptor Research, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ruimin Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Minjun Li
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jianhua Shen
- State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yechun Xu
- CAS Key Laboratory of Receptor Research, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
367
|
Barthels F, Marincola G, Marciniak T, Konhäuser M, Hammerschmidt S, Bierlmeier J, Distler U, Wich PR, Tenzer S, Schwarzer D, Ziebuhr W, Schirmeister T. Asymmetric Disulfanylbenzamides as Irreversible and Selective Inhibitors of Staphylococcus aureus Sortase A. ChemMedChem 2020; 15:839-850. [PMID: 32118357 PMCID: PMC7318353 DOI: 10.1002/cmdc.201900687] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/07/2020] [Indexed: 12/19/2022]
Abstract
Staphylococcus aureus is one of the most frequent causes of nosocomial and community-acquired infections, with drug-resistant strains being responsible for tens of thousands of deaths per year. S. aureus sortase A inhibitors are designed to interfere with virulence determinants. We have identified disulfanylbenzamides as a new class of potent inhibitors against sortase A that act by covalent modification of the active-site cysteine. A broad series of derivatives were synthesized to derive structure-activity relationships (SAR). In vitro and in silico methods allowed the experimentally observed binding affinities and selectivities to be rationalized. The most active compounds were found to have single-digit micromolar Ki values and caused up to a 66 % reduction of S. aureus fibrinogen attachment at an effective inhibitor concentration of 10 μM. This new molecule class exhibited minimal cytotoxicity, low bacterial growth inhibition and impaired sortase-mediated adherence of S. aureus cells.
Collapse
Affiliation(s)
- Fabian Barthels
- Institute for Pharmacy and BiochemistryJohannes-Gutenberg-University of MainzStaudinger Weg 555128MainzGermany
| | - Gabriella Marincola
- Institute for Molecular Infection BiologyJulius-Maximilians-University of WürzburgJosef-Schneider-Strasse 297080WürzburgGermany
| | - Tessa Marciniak
- Institute for Molecular Infection BiologyJulius-Maximilians-University of WürzburgJosef-Schneider-Strasse 297080WürzburgGermany
| | - Matthias Konhäuser
- Institute for Pharmacy and BiochemistryJohannes-Gutenberg-University of MainzStaudinger Weg 555128MainzGermany
| | - Stefan Hammerschmidt
- Institute for Pharmacy and BiochemistryJohannes-Gutenberg-University of MainzStaudinger Weg 555128MainzGermany
| | - Jan Bierlmeier
- Interfaculty Institute of BiochemistryEberhard-Karls-University of TübingenHoppe-Seyler-Strasse 472076TübingenGermany
| | - Ute Distler
- Institute for ImmunologyUniversity Medical CenterJohannes-Gutenberg-University of MainzLangenbeckstr. 155131MainzGermany
- Focus Program Translational Neuroscience (FTN)University Medical CenterLangenbeckstr. 155131MainzGermany
| | - Peter R. Wich
- Institute for Pharmacy and BiochemistryJohannes-Gutenberg-University of MainzStaudinger Weg 555128MainzGermany
- School of Chemical EngineeringUniversity of New South WalesScience and Engineering BuildingSydneyNSW 2052Australia
| | - Stefan Tenzer
- Institute for ImmunologyUniversity Medical CenterJohannes-Gutenberg-University of MainzLangenbeckstr. 155131MainzGermany
| | - Dirk Schwarzer
- Interfaculty Institute of BiochemistryEberhard-Karls-University of TübingenHoppe-Seyler-Strasse 472076TübingenGermany
| | - Wilma Ziebuhr
- Institute for Molecular Infection BiologyJulius-Maximilians-University of WürzburgJosef-Schneider-Strasse 297080WürzburgGermany
| | - Tanja Schirmeister
- Institute for Pharmacy and BiochemistryJohannes-Gutenberg-University of MainzStaudinger Weg 555128MainzGermany
| |
Collapse
|
368
|
Palazzesi F, Hermann MR, Grundl MA, Pautsch A, Seeliger D, Tautermann CS, Weber A. BIreactive: A Machine-Learning Model to Estimate Covalent Warhead Reactivity. J Chem Inf Model 2020; 60:2915-2923. [PMID: 32250627 DOI: 10.1021/acs.jcim.9b01058] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the past decade, the pharmaceutical industry has paid closer attention to covalent drugs. Differently from standard noncovalent drugs, these compounds can exhibit peculiar properties, such as higher potency or longer duration of target inhibition with a potentially lower dosage. These properties are mainly driven by the reactive functional group present in the compound, the so-called warhead that forms a covalent bond with a specific nucleophilic amino-acid on the target. In this work, we report the possibility to combine ab initio activation energies with machine-learning to estimate covalent compound intrinsic reactivity. The idea behind this approach is to have a precise estimation of the transition state barriers, and thus of the compound reactivity, but with the speed of a machine-learning algorithm. We call this method "BIreactive". Here, we demonstrate this approach on acrylamides and 2-chloroacetamides, two warhead classes that possess different reaction mechanisms. In combination with our recently implemented truncation algorithm, we also demonstrate the possibility to use BIreactive not only for fragments but also for lead-like molecules. The generic nature of this approach allows also the extension to several other warheads. The combination of these factors makes BIreactive a valuable tool for the covalent drug discovery process in a pharmaceutical context.
Collapse
Affiliation(s)
- Ferruccio Palazzesi
- Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorferstrasse 65, 88397 Biberach an der Riß, Germany
| | - Markus R Hermann
- Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorferstrasse 65, 88397 Biberach an der Riß, Germany
| | - Marc A Grundl
- Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorferstrasse 65, 88397 Biberach an der Riß, Germany
| | - Alexander Pautsch
- Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorferstrasse 65, 88397 Biberach an der Riß, Germany
| | - Daniel Seeliger
- Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorferstrasse 65, 88397 Biberach an der Riß, Germany
| | - Christofer S Tautermann
- Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorferstrasse 65, 88397 Biberach an der Riß, Germany
| | - Alexander Weber
- Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorferstrasse 65, 88397 Biberach an der Riß, Germany
| |
Collapse
|
369
|
Keeley A, Petri L, Ábrányi-Balogh P, Keserű GM. Covalent fragment libraries in drug discovery. Drug Discov Today 2020; 25:983-996. [PMID: 32298798 DOI: 10.1016/j.drudis.2020.03.016] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/07/2020] [Accepted: 03/23/2020] [Indexed: 12/20/2022]
Abstract
Targeted covalent inhibitors and chemical probes have become integral parts of drug discovery approaches. Given the advantages of fragment-based drug discovery, screening electrophilic fragments emerged as a promising alternative to discover and validate novel targets and to generate viable chemical starting points even for targets that are barely tractable. In this review, we present recent principles and considerations in the design of electrophilic fragment libraries from the selection of the appropriate covalent warhead through the design of the covalent fragment to the compilation of the library. We then summarize recent screening methodologies of covalent fragments against surrogate models, proteins, and the whole proteome, or living cells. Finally, we highlight recent drug discovery applications of covalent fragment libraries.
Collapse
Affiliation(s)
- Aaron Keeley
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
| | - László Petri
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
| | - Péter Ábrányi-Balogh
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
| | - György M Keserű
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar tudósok krt. 2, H-1117 Budapest, Hungary.
| |
Collapse
|
370
|
Cheng M, Guo C, Gross ML. The Application of Fluorine-Containing Reagents in Structural Proteomics. Angew Chem Int Ed Engl 2020; 59:5880-5889. [PMID: 31588625 PMCID: PMC7485648 DOI: 10.1002/anie.201907662] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Indexed: 01/01/2023]
Abstract
Structural proteomics refers to large-scale mapping of protein structures in order to understand the relationship between protein sequence, structure, and function. Chemical labeling, in combination with mass-spectrometry (MS) analysis, have emerged as powerful tools to enable a broad range of biological applications in structural proteomics. The key to success is a biocompatible reagent that modifies a protein without affecting its high-order structure. Fluorine, well-known to exert profound effects on the physical and chemical properties of reagents, should have an impact on structural proteomics. In this Minireview, we describe several fluorine-containing reagents that can be applied in structural proteomics. We organize their applications around four MS-based techniques: a) affinity labeling, b) activity-based protein profiling (ABPP), c) protein footprinting, and d) protein cross-linking. Our aim is to provide an overview of the research, development, and application of fluorine-containing reagents in protein structural studies.
Collapse
Affiliation(s)
- Ming Cheng
- Department of Chemistry, Washington University in St Louis, St Louis, MO 63130
| | - Chunyang Guo
- Department of Chemistry, Washington University in St Louis, St Louis, MO 63130
| | - Michael L Gross
- Department of Chemistry, Washington University in St Louis, St Louis, MO 63130
| |
Collapse
|
371
|
New Cysteine Protease Inhibitors: Electrophilic (Het)arenes and Unexpected Prodrug Identification for the Trypanosoma Protease Rhodesain. Molecules 2020; 25:molecules25061451. [PMID: 32210166 PMCID: PMC7145299 DOI: 10.3390/molecules25061451] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/07/2020] [Accepted: 03/13/2020] [Indexed: 01/05/2023] Open
Abstract
Electrophilic (het)arenes can undergo reactions with nucleophiles yielding π- or Meisenheimer (σ-) complexes or the products of the SNAr addition/elimination reactions. Such building blocks have only rarely been employed for the design of enzyme inhibitors. Herein, we demonstrate the combination of a peptidic recognition sequence with such electrophilic (het)arenes to generate highly active inhibitors of disease-relevant proteases. We further elucidate an unexpected mode of action for the trypanosomal protease rhodesain using NMR spectroscopy and mass spectrometry, enzyme kinetics and various types of simulations. After hydrolysis of an ester function in the recognition sequence of a weakly active prodrug inhibitor, the liberated carboxylic acid represents a highly potent inhibitor of rhodesain (Ki = 4.0 nM). The simulations indicate that, after the cleavage of the ester, the carboxylic acid leaves the active site and re-binds to the enzyme in an orientation that allows the formation of a very stable π-complex between the catalytic dyad (Cys-25/His-162) of rhodesain and the electrophilic aromatic moiety. The reversible inhibition mode results because the SNAr reaction, which is found in an alkaline solvent containing a low molecular weight thiol, is hindered within the enzyme due to the presence of the positively charged imidazolium ring of His-162. Comparisons between measured and calculated NMR shifts support this interpretation.
Collapse
|
372
|
Small-molecule covalent bond formation at tyrosine creates a binding site and inhibits activation of Ral GTPases. Proc Natl Acad Sci U S A 2020; 117:7131-7139. [PMID: 32179690 DOI: 10.1073/pnas.1913654117] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Ral (Ras-like) GTPases are directly activated by oncogenic Ras GTPases. Mutant K-Ras (G12C) has enabled the development of covalent K-Ras inhibitors currently in clinical trials. However, Ral, and the overwhelming majority of mutant oncogenic K-Ras, are devoid of a druggable pocket and lack an accessible cysteine for the development of a covalent inhibitor. Here, we report that covalent bond formation by an aryl sulfonyl fluoride electrophile at a tyrosine residue (Tyr-82) inhibits guanine exchange factor Rgl2-mediated nucleotide exchange of Ral GTPase. A high-resolution 1.18-Å X-ray cocrystal structure shows that the compound binds to a well-defined binding site in RalA as a result of a switch II loop conformational change. The structure, along with additional high-resolution crystal structures of several analogs in complex with RalA, confirm the importance of key hydrogen bond anchors between compound sulfone oxygen atoms and Ral backbone nitrogen atoms. Our discovery of a pocket with features found on known druggable sites and covalent modification of a bystander tyrosine residue present in Ral and Ras GTPases provide a strategy that could lead to therapeutic agent targeting oncogenic Ras mutants that are devoid of a cysteine nucleophile.
Collapse
|
373
|
Abstract
The use of an acetylene (ethynyl) group in medicinal chemistry coincides with the launch of the Journal of Medicinal Chemistry in 1959. Since then, the acetylene group has been broadly exploited in drug discovery and development. As a result, it has become recognized as a privileged structural feature for targeting a wide range of therapeutic target proteins, including MAO, tyrosine kinases, BACE1, steroid receptors, mGlu5 receptors, FFA1/GPR40, and HIV-1 RT. Furthermore, a terminal alkyne functionality is frequently introduced in chemical biology probes as a click handle to identify molecular targets and to assess target engagement. This Perspective is divided into three parts encompassing: (1) the physicochemical properties of the ethynyl group, (2) the advantages and disadvantages of the ethynyl group in medicinal chemistry, and (3) the impact of the ethynyl group on chemical biology approaches.
Collapse
Affiliation(s)
- Tanaji T Talele
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York 11439, United States
| |
Collapse
|
374
|
Petri L, Ábrányi-Balogh P, Varga PR, Imre T, Keserű GM. Comparative reactivity analysis of small-molecule thiol surrogates. Bioorg Med Chem 2020; 28:115357. [PMID: 32081630 DOI: 10.1016/j.bmc.2020.115357] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/21/2020] [Accepted: 01/30/2020] [Indexed: 12/12/2022]
Abstract
Targeted covalent inhibitors represent an increasingly popular approach to modulate challenging drug targets. Since covalent and non-covalent interactions are both contributing to the affinity of these compounds, evaluation of their reactivity is a key-step to find feasible warheads. There are well-established HPLC- and NMR-based kinetic assays to tackle this task, however, they use a variety of cysteine-surrogates including cysteamine, cysteine or acetyl-cysteine and GSH. The diverse nature of the thiol sources often makes the results incomparable that prevents compiling a comprehensive knowledge base for the design of covalent inhibitors. To evaluate kinetic measurements from different sources we performed a comparative analysis of the different thiol surrogates against a designed set of electrophilic fragments equipped with a range of warheads. Our study included seven different thiol models and 13 warheads resulting in a reactivity matrix analysed thoroughly. We found that the reactivity profile might be significantly different for various thiol models. Comparing the different warheads, we concluded that - in addition to its human relevance - glutathione (GSH) provided the best estimate of reactivity with highest number of true positives identified.
Collapse
Affiliation(s)
- László Petri
- Research Centre for Natural Sciences, Medicinal Chemistry Research Group, H-1117 Budapest, Magyar tudósok krt 2, Hungary
| | - Péter Ábrányi-Balogh
- Research Centre for Natural Sciences, Medicinal Chemistry Research Group, H-1117 Budapest, Magyar tudósok krt 2, Hungary
| | - Petra Regina Varga
- Research Centre for Natural Sciences, Medicinal Chemistry Research Group, H-1117 Budapest, Magyar tudósok krt 2, Hungary
| | - Tímea Imre
- Research Centre for Natural Sciences, MS Metabolomics Research Group, H-1117 Budapest, Magyar tudósok krt 2, Hungary
| | - György Miklós Keserű
- Research Centre for Natural Sciences, Medicinal Chemistry Research Group, H-1117 Budapest, Magyar tudósok krt 2, Hungary.
| |
Collapse
|
375
|
Wang R, Chen Y, Shu M, Zhao W, Tao M, Du C, Fu X, Li A, Lin Z. AuCl 3 -Catalyzed Ring-Closing Carbonyl-Olefin Metathesis. Chemistry 2020; 26:1941-1946. [PMID: 31867760 DOI: 10.1002/chem.201905199] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 12/19/2019] [Indexed: 11/05/2022]
Abstract
Compared with the ripeness of olefin metathesis, exploration of the construction of carbon-carbon double bonds through the catalytic carbonyl-olefin metathesis reaction remains stagnant and has received scant attention. Herein, a highly efficient AuCl3 -catalyzed intramolecular ring-closing carbonyl-olefin metathesis reaction is described. This method features easily accessible starting materials, simple operation, good functional-group tolerance and short reaction times, and provides the target cyclopentenes, polycycles, benzocarbocycles, and N-heterocycle derivatives in good to excellent yields.
Collapse
Affiliation(s)
- Rui Wang
- School of Pharmacy & Bioengineering, Chongqing University of Technology, 69 Red Avenue, Chongqing, 400054, China
| | - Yi Chen
- School of Pharmacy & Bioengineering, Chongqing University of Technology, 69 Red Avenue, Chongqing, 400054, China
| | - Mao Shu
- School of Pharmacy & Bioengineering, Chongqing University of Technology, 69 Red Avenue, Chongqing, 400054, China
| | - Wenwen Zhao
- School of Pharmacy & Bioengineering, Chongqing University of Technology, 69 Red Avenue, Chongqing, 400054, China
| | - Maoling Tao
- School of Pharmacy & Bioengineering, Chongqing University of Technology, 69 Red Avenue, Chongqing, 400054, China
| | - Chao Du
- School of Pharmacy & Bioengineering, Chongqing University of Technology, 69 Red Avenue, Chongqing, 400054, China
| | - Xiaoya Fu
- School of Pharmacy & Bioengineering, Chongqing University of Technology, 69 Red Avenue, Chongqing, 400054, China
| | - Ao Li
- School of Pharmacy & Bioengineering, Chongqing University of Technology, 69 Red Avenue, Chongqing, 400054, China
| | - Zhihua Lin
- School of Pharmacy & Bioengineering, Chongqing University of Technology, 69 Red Avenue, Chongqing, 400054, China
| |
Collapse
|
376
|
Dalton SE, Campos S. Covalent Small Molecules as Enabling Platforms for Drug Discovery. Chembiochem 2020; 21:1080-1100. [DOI: 10.1002/cbic.201900674] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Samuel E. Dalton
- Astex Pharmaceuticals 436 Cambridge Science Park Milton Road Cambridge CB4 0QA UK
| | - Sebastien Campos
- PharmaronDrug Discovery Services Europe Hertford Road Hoddesdon Hertfordshire EN11 9BU UK
| |
Collapse
|
377
|
Cuesta A, Wan X, Burlingame AL, Taunton J. Ligand Conformational Bias Drives Enantioselective Modification of a Surface-Exposed Lysine on Hsp90. J Am Chem Soc 2020; 142:3392-3400. [DOI: 10.1021/jacs.9b09684] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Adolfo Cuesta
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California 94158, United States
| | - Xiaobo Wan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California 94158, United States
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158, United States
| | - Alma L. Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158, United States
| | - Jack Taunton
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California 94158, United States
| |
Collapse
|
378
|
|
379
|
Lodola A, Callegari D, Scalvini L, Rivara S, Mor M. Design and SAR Analysis of Covalent Inhibitors Driven by Hybrid QM/MM Simulations. Methods Mol Biol 2020; 2114:307-337. [PMID: 32016901 DOI: 10.1007/978-1-0716-0282-9_19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Quantum mechanics/molecular mechanics (QM/MM) hybrid technique is emerging as a reliable computational method to investigate and characterize chemical reactions occurring in enzymes. From a drug discovery perspective, a thorough understanding of enzyme catalysis appears pivotal to assist the design of inhibitors able to covalently bind one of the residues belonging to the enzyme catalytic machinery. Thanks to the current advances in computer power, and the availability of more efficient algorithms for QM-based simulations, the use of QM/MM methodology is becoming a viable option in the field of covalent inhibitor design. In the present review, we summarized our experience in the field of QM/MM simulations applied to drug design problems which involved the optimization of agents working on two well-known drug targets, namely fatty acid amide hydrolase (FAAH) and epidermal growth factor receptor (EGFR). In this context, QM/MM simulations gave valuable information in terms of geometry (i.e., of transition states and metastable intermediates) and reaction energetics that allowed to correctly predict inhibitor binding orientation and substituent effect on enzyme inhibition. What is more, enzyme reaction modelling with QM/MM provided insights that were translated into the synthesis of new covalent inhibitor featured by a unique combination of intrinsic reactivity, on-target activity, and selectivity.
Collapse
Affiliation(s)
- Alessio Lodola
- Drug Design and Discovery Group, Department of Food and Drug, University of Parma, Parma, Italy.
| | - Donatella Callegari
- Drug Design and Discovery Group, Department of Food and Drug, University of Parma, Parma, Italy
| | - Laura Scalvini
- Drug Design and Discovery Group, Department of Food and Drug, University of Parma, Parma, Italy
| | - Silvia Rivara
- Drug Design and Discovery Group, Department of Food and Drug, University of Parma, Parma, Italy
| | - Marco Mor
- Drug Design and Discovery Group, Department of Food and Drug, University of Parma, Parma, Italy
| |
Collapse
|
380
|
Abstract
Intrinsically disordered proteins (IDPs) play important roles in the regulation of cellular function and in disease, and thus they represent an important group of therapeutic targets. Yet, members of this "disorderome" have not yet been successfully targeted by drugs, primarily because traditional design principles cannot be applied to their highly dynamic, heterogeneous structural states. Binders developed against IDPs so far suffer from very weak binding and inability to act in a cellular context. Here, we describe a possible generic method for the targeting of IDPs via covalent modification, which could entail specific and strong binding and inhibitory potential, making such "warheads" reasonable starting points of drug-development efforts. We demonstrate this principle by addressing the cysteine-specific covalent modification of calpastatin, the IDP inhibitor of the calcium-dependent cysteine protease calpain. We describe the protocol for monitoring the covalent modification of the inhibitor, measuring the Ki of its inhibition and comparing it to the Kd of its interaction with the enzyme. Our premise is that the underlying principles can be easily adapted to screen for molecules targeting other, disease-related, IDPs in the future.
Collapse
|
381
|
Abdeldayem A, Raouf YS, Constantinescu SN, Moriggl R, Gunning PT. Advances in covalent kinase inhibitors. Chem Soc Rev 2020; 49:2617-2687. [DOI: 10.1039/c9cs00720b] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This comprehensive review details recent advances, challenges and innovations in covalent kinase inhibition within a 10 year period (2007–2018).
Collapse
Affiliation(s)
- Ayah Abdeldayem
- Department of Chemical & Physical Sciences
- University of Toronto
- Mississauga
- Canada
- Department of Chemistry
| | - Yasir S. Raouf
- Department of Chemical & Physical Sciences
- University of Toronto
- Mississauga
- Canada
- Department of Chemistry
| | | | - Richard Moriggl
- Institute of Animal Breeding and Genetics
- University of Veterinary Medicine
- 1210 Vienna
- Austria
| | - Patrick T. Gunning
- Department of Chemical & Physical Sciences
- University of Toronto
- Mississauga
- Canada
- Department of Chemistry
| |
Collapse
|
382
|
Bosc D, Camberlein V, Gealageas R, Castillo-Aguilera O, Deprez B, Deprez-Poulain R. Kinetic Target-Guided Synthesis: Reaching the Age of Maturity. J Med Chem 2019; 63:3817-3833. [PMID: 31820982 DOI: 10.1021/acs.jmedchem.9b01183] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Kinetic target-guided synthesis (KTGS) is an original discovery strategy allowing a target to catalyze the irreversible synthesis of its own ligands from a pool of reagents. Although pioneered almost two decades ago, it only recently proved its usefulness in medicinal chemistry, as exemplified by the increasing number of protein targets used, the wider range of target and pocket types, and the diversity of therapeutic areas explored. In recent years, two new leads for in vivo studies were released. Amidations and multicomponent reactions expanded the armamentarium of reactions beyond triazole formation and two new examples of in cellulo KTGS were also disclosed. Herein, we analyze the origins and the chemical space of both KTGS ligands and warhead-bearing reagents. We review the KTGS timeline focusing on recent cases in order to give medicinal chemists the full scope of this strategy which has great potential for hit discovery and hit or lead optimization.
Collapse
Affiliation(s)
- Damien Bosc
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000 Lille, France
| | - Virgyl Camberlein
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000 Lille, France
| | - Ronan Gealageas
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000 Lille, France
| | - Omar Castillo-Aguilera
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000 Lille, France
| | - Benoit Deprez
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000 Lille, France
| | - Rebecca Deprez-Poulain
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000 Lille, France.,Institut Universitaire de France, F- 75005 Paris, France
| |
Collapse
|
383
|
Zhou S, Pan J, Davis KM, Schaperdoth I, Wang B, Boal AK, Krebs C, Bollinger JM. Steric Enforcement of cis-Epoxide Formation in the Radical C-O-Coupling Reaction by Which ( S)-2-Hydroxypropylphosphonate Epoxidase (HppE) Produces Fosfomycin. J Am Chem Soc 2019; 141:20397-20406. [PMID: 31769979 DOI: 10.1021/jacs.9b10974] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
(S)-2-Hydroxypropylphosphonate [(S)-2-HPP, 1] epoxidase (HppE) reduces H2O2 at its nonheme-iron cofactor to install the oxirane "warhead" of the antibiotic fosfomycin. The net replacement of the C1 pro-R hydrogen of 1 by its C2 oxygen, with inversion of configuration at C1, yields the cis-epoxide of the drug [(1R,2S)-epoxypropylphosphonic acid (cis-Fos, 2)]. Here we show that HppE achieves ∼95% selectivity for C1 inversion and cis-epoxide formation via steric guidance of a radical-coupling mechanism. Published structures of the HppE·FeII·1 and HppE·ZnII·2 complexes reveal distinct pockets for C3 of the substrate and product and identify four hydrophobic residues-Leu120, Leu144, Phe182, and Leu193-close to C3 in one of the complexes. Replacement of Leu193 in the substrate C3 pocket with the bulkier Phe enhances stereoselectivity (cis:trans ∼99:1), whereas the Leu120Phe substitution in the product C3 pocket diminishes it (∼82:18). Retention of C1 configuration and trans-epoxide formation become predominant with the bulk-reducing Phe182Ala substitution in the substrate C3 pocket (∼13:87), trifluorination of C3 (∼23:77), or both (∼1:99). The effect of C3 trifluorination is counteracted by the more constrained substrate C3 pockets in the Leu193Phe (∼56:44) and Leu144Phe/Leu193Phe (∼90:10) variants. The ability of HppE to epoxidize substrate analogues bearing halogens at C3, C1, or both is inconsistent with a published hypothesis of polar cyclization via a C1 carbocation. Rather, specific enzyme-substrate contacts drive inversion of the C1 radical-as proposed in a recent computational study-to direct formation of the more potently antibacterial cis-epoxide by radicaloid C-O coupling.
Collapse
Affiliation(s)
- Shengbin Zhou
- Department of Chemistry and Department of Biochemistry & Molecular Biology , The Pennsylvania State University , University Park , Pennsylvania 16802 , United States
| | - Juan Pan
- Department of Chemistry and Department of Biochemistry & Molecular Biology , The Pennsylvania State University , University Park , Pennsylvania 16802 , United States
| | - Katherine M Davis
- Department of Chemistry and Department of Biochemistry & Molecular Biology , The Pennsylvania State University , University Park , Pennsylvania 16802 , United States
| | - Irene Schaperdoth
- Department of Chemistry and Department of Biochemistry & Molecular Biology , The Pennsylvania State University , University Park , Pennsylvania 16802 , United States
| | - Bo Wang
- Department of Chemistry and Department of Biochemistry & Molecular Biology , The Pennsylvania State University , University Park , Pennsylvania 16802 , United States
| | - Amie K Boal
- Department of Chemistry and Department of Biochemistry & Molecular Biology , The Pennsylvania State University , University Park , Pennsylvania 16802 , United States
| | - Carsten Krebs
- Department of Chemistry and Department of Biochemistry & Molecular Biology , The Pennsylvania State University , University Park , Pennsylvania 16802 , United States
| | - J Martin Bollinger
- Department of Chemistry and Department of Biochemistry & Molecular Biology , The Pennsylvania State University , University Park , Pennsylvania 16802 , United States
| |
Collapse
|
384
|
Baska F, Sipos A, Őrfi Z, Nemes Z, Dobos J, Szántai-Kis C, Szabó E, Szénási G, Dézsi L, Hamar P, Cserepes MT, Tóvári J, Garamvölgyi R, Krekó M, Őrfi L. Discovery and development of extreme selective inhibitors of the ITD and D835Y mutant FLT3 kinases. Eur J Med Chem 2019; 184:111710. [DOI: 10.1016/j.ejmech.2019.111710] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/02/2019] [Accepted: 09/16/2019] [Indexed: 10/25/2022]
|
385
|
Moku B, Fang WY, Leng J, Li L, Zha GF, Rakesh KP, Qin HL. Rh-Catalyzed Highly Enantioselective Synthesis of Aliphatic Sulfonyl Fluorides. iScience 2019; 21:695-705. [PMID: 31733515 PMCID: PMC6889689 DOI: 10.1016/j.isci.2019.10.051] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 10/14/2019] [Accepted: 10/24/2019] [Indexed: 11/25/2022] Open
Abstract
Rh-catalyzed, highly enantioselective (up to 99.8% ee) synthesis of aliphatic sulfonyl fluorides was accomplished. This protocol provides a portal to a class of novel 2-aryl substituted chiral sulfonyl fluorides, which are otherwise extremely difficult to access. This asymmetric synthesis has the feature of mild conditions, excellent functional group compatibility, and wide substrate scope (51 examples) generating a wide array of structurally unique chiral β-arylated sulfonyl fluorides for sulfur(VI) fluoride exchange (SuFEx) click reaction and drug discovery.
Collapse
Affiliation(s)
- Balakrishna Moku
- State Key Laboratory of Silicate Materials for Architectures, and School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan 430070, P. R. China
| | - Wan-Yin Fang
- State Key Laboratory of Silicate Materials for Architectures, and School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan 430070, P. R. China
| | - Jing Leng
- State Key Laboratory of Silicate Materials for Architectures, and School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan 430070, P. R. China
| | - Linxian Li
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institute, Hong Kong, China
| | - Gao-Feng Zha
- State Key Laboratory of Silicate Materials for Architectures, and School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan 430070, P. R. China; Ming Wai Lau Centre for Reparative Medicine, Karolinska Institute, Hong Kong, China
| | - K P Rakesh
- State Key Laboratory of Silicate Materials for Architectures, and School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan 430070, P. R. China
| | - Hua-Li Qin
- State Key Laboratory of Silicate Materials for Architectures, and School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan 430070, P. R. China.
| |
Collapse
|
386
|
Voice A, Tresadern G, van Vlijmen H, Mulholland A. Limitations of Ligand-Only Approaches for Predicting the Reactivity of Covalent Inhibitors. J Chem Inf Model 2019; 59:4220-4227. [PMID: 31498988 DOI: 10.1021/acs.jcim.9b00404] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Covalent inhibition has undergone a resurgence and is an important modern-day drug design and chemical biology approach. To avoid off-target interactions and to fine-tune reactivity, the ability to accurately predict reactivity is vitally important for the design and development of safer and more effective covalent drugs. Several ligand-only metrics have been proposed that promise quick and simple ways of determining covalent reactivity. In particular, we examine proton affinity and reaction energies calculated with the density functional B3LYP-D3/6-311+G**//B3LYP-D3/6-31G* method to assess the reactivity of a series of α,β-unsaturated carbonyl compounds that form covalent adducts with cysteine. We demonstrate that while these metrics correlate well with experiment for a diverse range of small reactive molecules these approaches fail for predicting the reactivity of drug-like compounds. We conclude that ligand-only metrics such as proton affinity and reaction energies do not capture determinants of reactivity in situ and fail to account for important factors such as conformation, solvation, and intermolecular interactions.
Collapse
Affiliation(s)
- Angus Voice
- Centre for Computational Chemistry, School of Chemistry , University of Bristol , Bristol BS8 1TS , United Kingdom
| | - Gary Tresadern
- Computational Chemistry, Janssen Research & Development , Janssen Pharmaceutica N. V. , Turnhoutseweg 30 , B-2340 Beerse , Belgium
| | - Herman van Vlijmen
- Computational Chemistry, Janssen Research & Development , Janssen Pharmaceutica N. V. , Turnhoutseweg 30 , B-2340 Beerse , Belgium
| | - Adrian Mulholland
- Centre for Computational Chemistry, School of Chemistry , University of Bristol , Bristol BS8 1TS , United Kingdom
| |
Collapse
|
387
|
Recent Advances in Selective and Irreversible Covalent Ligand Development and Validation. Cell Chem Biol 2019; 26:1486-1500. [PMID: 31631011 DOI: 10.1016/j.chembiol.2019.09.012] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 08/06/2019] [Accepted: 09/26/2019] [Indexed: 12/20/2022]
Abstract
Some of the most widely used drugs, such as aspirin and penicillin, are covalent drugs. Covalent binding can improve potency, selectivity, and duration of the effects, but the intrinsic reactivity represents a potential liability and may result in idiosyncratic toxicity. For decades, the cons were believed to outweigh the pros, and covalent targeting was deprioritized in drug discovery. Recently, several covalent inhibitors have been approved for cancer treatment, thus rebooting the field. In this review, we briefly reflect on the history of selective covalent targeting, and provide a comprehensive overview of emerging developments from a chemical biology stand-point. Our discussion will reflect on efforts to validate irreversible covalent ligands, expand the scope of targets, and discover new ligands and warheads. We conclude with a brief commentary of remaining limitations and emerging opportunities in selective covalent targeting.
Collapse
|
388
|
Baggio C, Udompholkul P, Gambini L, Salem AF, Jossart J, Perry JJP, Pellecchia M. Aryl-fluorosulfate-based Lysine Covalent Pan-Inhibitors of Apoptosis Protein (IAP) Antagonists with Cellular Efficacy. J Med Chem 2019; 62:9188-9200. [PMID: 31550155 DOI: 10.1021/acs.jmedchem.9b01108] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We have recently investigated the reactivity of aryl-fluorosulfates as warheads to form covalent adducts with Lys, Tyr, and His residues. However, the rate of reaction of aryl-fluorosulfates seemed relatively slow, putting into question their effectiveness to form covalent adducts in cell. Unlike the previously reported agents that targeted a relatively remote Lys residue with respect to the target's binding site, the current agents were designed to more directly juxtapose an aryl-fluorosulfate with a Lys residue that is located within the binding pocket of the BIR3 domain of X-linked inhibitor of apoptosis protein (XIAP). We found that such new agents can effectively and rapidly form a covalent adduct with XIAP-BIR3 in vitro and in cell, approaching the rate of reaction, cellular permeability, and stability that are similar to what attained by acrylamides when targeting Cys residues. Our studies further validate aryl-fluorosulfates as valuable Lys-targeting electrophiles, for the design of inhibitors of both enzymes and protein-protein interactions.
Collapse
|
389
|
Moku B, Fang WY, Leng J, Kantchev EAB, Qin HL. Rh(I)–Diene-Catalyzed Addition of (Hetero)aryl Functionality to 1,3-Dienylsulfonyl Fluorides Achieving Exclusive Regioselectivity and High Enantioselectivity: Generality and Mechanism. ACS Catal 2019. [DOI: 10.1021/acscatal.9b03640] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Balakrishna Moku
- State Key Laboratory of Silicate Materials for Architectures; and School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan 430070, China
| | - Wan-Yin Fang
- State Key Laboratory of Silicate Materials for Architectures; and School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan 430070, China
| | - Jing Leng
- State Key Laboratory of Silicate Materials for Architectures; and School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan 430070, China
| | - Eric Assen B. Kantchev
- State Key Laboratory of Silicate Materials for Architectures; and School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan 430070, China
| | - Hua-Li Qin
- State Key Laboratory of Silicate Materials for Architectures; and School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan 430070, China
| |
Collapse
|
390
|
Mukherjee H, Su N, Belmonte MA, Hargreaves D, Patel J, Tentarelli S, Aquila B, Grimster NP. Discovery and optimization of covalent Bcl-xL antagonists. Bioorg Med Chem Lett 2019; 29:126682. [PMID: 31606346 DOI: 10.1016/j.bmcl.2019.126682] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 12/12/2022]
Abstract
Over the last ten years, targeted covalent inhibition has become a key discipline within medicinal chemistry research, most notably in the development of oncology therapeutics. One area where this approach is underrepresented, however, is in targeting protein-protein interactions. This is primarily because these hydrophobic interfaces lack appropriately located cysteine residues to allow for standard conjugate addition chemistry. Herein, we report our development of the first covalent inhibitors of the antiapoptotic protein B-cell lymphoma extra-large (Bcl-xL), utilizing a sulfonyl fluoride (SF) warhead to selectively covalently modify tyrosine 101 of the BH3 domain-binding groove. These compounds display time-dependent inhibition in a biochemical assay and are cellularly active (U266B1). In addition, compound 7 was further elaborated to generate a chemical-biology probe molecule, which may find utility in understanding the intricacies of Bcl-xL biology.
Collapse
Affiliation(s)
| | - Nancy Su
- Discovery Sciences, R&D, AstraZeneca, Waltham, USA
| | | | | | - Joe Patel
- Discovery Sciences, R&D, AstraZeneca, Waltham, USA
| | | | | | | |
Collapse
|
391
|
Awoonor-Williams E, Isley WC, Dale SG, Johnson ER, Yu H, Becke AD, Roux B, Rowley CN. Quantum Chemical Methods for Modeling Covalent Modification of Biological Thiols. J Comput Chem 2019; 41:427-438. [PMID: 31512279 DOI: 10.1002/jcc.26064] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/24/2019] [Accepted: 08/16/2019] [Indexed: 02/06/2023]
Abstract
Targeted covalent inhibitor drugs require computational methods that go beyond simple molecular-mechanical force fields in order to model the chemical reactions that occur when they bind to their targets. Here, several semiempirical and density-functional theory (DFT) methods are assessed for their ability to describe the potential energy surface and reaction energies of the covalent modification of a thiol by an electrophile. Functionals such as PBE and B3LYP fail to predict a stable enolate intermediate. This is largely due to delocalization error, which spuriously stabilizes the prereaction complex, in which excess electron density is transferred from the thiolate to the electrophile. Functionals with a high-exact exchange component, range-separated DFT functionals, and variationally optimized exact exchange (i.e., the LC-B05minV functional) correct this issue to various degrees. The large gradient behavior of the exchange enhancement factor is also found to significantly affect the results, leading to the improved performance of PBE0. While ωB97X-D and M06-2X were reasonably accurate, no method provided quantitative accuracy for all three electrophiles, making this a very strenuous test of functional performance. Additionally, one drawback of M06-2X was that molecular dynamics (MD) simulations using this functional were only stable if a fine integration grid was used. The low-cost semiempirical methods, PM3, AM1, and PM7, provide a qualitatively correct description of the reaction mechanism, although the energetics is not quantitatively reliable. As a proof of concept, the potential of mean force for the addition of methylthiolate to methylvinyl ketone was calculated using quantum mechanical/molecular mechanical MD in an explicit polarizable aqueous solvent. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ernest Awoonor-Williams
- Department of Chemistry, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| | - William C Isley
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois
| | - Stephen G Dale
- Research School of Chemistry, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Erin R Johnson
- Department of Chemistry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Haibo Yu
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, New South Wales, Australia
| | - Axel D Becke
- Department of Chemistry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Benoît Roux
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois
| | - Christopher N Rowley
- Department of Chemistry, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| |
Collapse
|
392
|
Leng J, Alharbi NS, Qin HL. Construction of α-(Hetero)aryl Ethenesulfonyl Fluorides for SuFEx Click Chemistry. European J Org Chem 2019. [DOI: 10.1002/ejoc.201901106] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Jing Leng
- State Key Laboratory of Silicate Materials for Architectures; and; School of Chemistry, Chemical Engineering and Life Science; Wuhan University of Technology; 430070 Wuhan Hubei Province People's Republic of China
| | - Njud S. Alharbi
- Biotechnology Research group; Deportment of Biological Sciences; Faculty of Science; King Abdulaziz University; Jeddah Saudi Arabia
| | - Hua-Li Qin
- State Key Laboratory of Silicate Materials for Architectures; and; School of Chemistry, Chemical Engineering and Life Science; Wuhan University of Technology; 430070 Wuhan Hubei Province People's Republic of China
| |
Collapse
|
393
|
Plescia J, De Cesco S, Patrascu MB, Kurian J, Di Trani J, Dufresne C, Wahba AS, Janmamode N, Mittermaier AK, Moitessier N. Integrated Synthetic, Biophysical, and Computational Investigations of Covalent Inhibitors of Prolyl Oligopeptidase and Fibroblast Activation Protein α. J Med Chem 2019; 62:7874-7884. [PMID: 31393718 DOI: 10.1021/acs.jmedchem.9b00642] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Jessica Plescia
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec H3A 0B8, Canada
| | - Stéphane De Cesco
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec H3A 0B8, Canada
| | - Mihai Burai Patrascu
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec H3A 0B8, Canada
| | - Jerry Kurian
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec H3A 0B8, Canada
| | - Justin Di Trani
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec H3A 0B8, Canada
| | - Caroline Dufresne
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec H3A 0B8, Canada
| | - Alexander S. Wahba
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec H3A 0B8, Canada
| | - Naëla Janmamode
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec H3A 0B8, Canada
| | - Anthony K. Mittermaier
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec H3A 0B8, Canada
| | - Nicolas Moitessier
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec H3A 0B8, Canada
| |
Collapse
|
394
|
Cianni L, Feldmann CW, Gilberg E, Gütschow M, Juliano L, Leitão A, Bajorath J, Montanari CA. Can Cysteine Protease Cross-Class Inhibitors Achieve Selectivity? J Med Chem 2019; 62:10497-10525. [DOI: 10.1021/acs.jmedchem.9b00683] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Lorenzo Cianni
- Medicinal Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, Avenue Trabalhador Sancarlense, 400, 23566-590 São Carlos, SP, Brazil
- Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität, Endenicher Allee 19c, D-53115 Bonn, Germany
| | - Christian Wolfgang Feldmann
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität, Endenicher Allee 19c, D-53115 Bonn, Germany
| | - Erik Gilberg
- Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität, Endenicher Allee 19c, D-53115 Bonn, Germany
| | - Michael Gütschow
- Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Luiz Juliano
- A. C. Camargo Cancer Center and São Paulo Medical School of Federal University of São Paulo, Rua Professor Antônio Prudente, 211, 01509-010 São Paulo, SP, Brazil
| | - Andrei Leitão
- Medicinal Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, Avenue Trabalhador Sancarlense, 400, 23566-590 São Carlos, SP, Brazil
| | - Jürgen Bajorath
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität, Endenicher Allee 19c, D-53115 Bonn, Germany
| | - Carlos A. Montanari
- Medicinal Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, Avenue Trabalhador Sancarlense, 400, 23566-590 São Carlos, SP, Brazil
| |
Collapse
|
395
|
Wørmer GJ, Hansen BK, Palmfeldt J, Poulsen TB. A Cyclopropene Electrophile that Targets Glutathione S‐Transferase Omega‐1 in Cells. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201907520] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Gustav J. Wørmer
- Department of Chemistry Aarhus University Langelandsgade 140 8000 Aarhus C Denmark
| | - Bente K. Hansen
- Department of Chemistry Aarhus University Langelandsgade 140 8000 Aarhus C Denmark
| | - Johan Palmfeldt
- Department of Clinical Medicine—Research Unit for Molecular Medicine Aarhus University hospital Palle Juul-Jensens Boulevard 82 8200 Aarhus N Denmark
| | - Thomas B. Poulsen
- Department of Chemistry Aarhus University Langelandsgade 140 8000 Aarhus C Denmark
| |
Collapse
|
396
|
Wørmer GJ, Hansen BK, Palmfeldt J, Poulsen TB. A Cyclopropene Electrophile that Targets Glutathione S‐Transferase Omega‐1 in Cells. Angew Chem Int Ed Engl 2019; 58:11918-11922. [DOI: 10.1002/anie.201907520] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Gustav J. Wørmer
- Department of Chemistry Aarhus University Langelandsgade 140 8000 Aarhus C Denmark
| | - Bente K. Hansen
- Department of Chemistry Aarhus University Langelandsgade 140 8000 Aarhus C Denmark
| | - Johan Palmfeldt
- Department of Clinical Medicine—Research Unit for Molecular Medicine Aarhus University hospital Palle Juul-Jensens Boulevard 82 8200 Aarhus N Denmark
| | - Thomas B. Poulsen
- Department of Chemistry Aarhus University Langelandsgade 140 8000 Aarhus C Denmark
| |
Collapse
|
397
|
Palazzesi F, Grundl MA, Pautsch A, Weber A, Tautermann CS. A Fast Ab Initio Predictor Tool for Covalent Reactivity Estimation of Acrylamides. J Chem Inf Model 2019; 59:3565-3571. [PMID: 31246457 DOI: 10.1021/acs.jcim.9b00316] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Thanks to their unique mode of action, covalent drugs represent an exceptional opportunity for drug design. After binding to a biologically relevant target system, covalent compounds form a reversible or irreversible covalent bond with a nucleophilic amino acid. Due to the inherently large binding energy of a covalent bond, covalent binders exhibit higher potencies and thus allow potentially lower drug dosages. However, a proper balancing of compound reactivity is key for the design of covalent binders, to achieve high levels of target inhibition while minimizing promiscuous covalent binding to nontarget proteins. In this work, we demonstrated the possibility to apply the electrophilicity index concept to estimate covalent compound reactivity. We tested this approach on acrylamides, one of the most prominent classes of covalent warheads. Our study clearly demonstrated that, for compounds with molecular weight (MW) below 250 Da, the electrophilicity index can be directly used to estimate compound reactivity. On the other hand, for leadlike molecules (MW > 250 Da) we implemented a new truncation algorithm that has to be applied before reactivity calculations. This algorithm can ensure the localization of HOMO/LUMO orbitals on the compound warhead and thus a correct estimation of its reactivity. Our results also indicate that caution should be used when employing the electrophilicity index to estimate the reactivity of nonterminal acrylamides. The nonparametric nature of this method and its reasonable computational cost make it a suitable tool to support covalent drug design.
Collapse
Affiliation(s)
- Ferruccio Palazzesi
- Medicinal Chemistry , Boehringer Ingelheim Pharma GmbH & Co. KG , Birkendorfer Strasse 65 , 88397 Biberach an der Riss , Germany
| | - Marc A Grundl
- Medicinal Chemistry , Boehringer Ingelheim Pharma GmbH & Co. KG , Birkendorfer Strasse 65 , 88397 Biberach an der Riss , Germany
| | - Alexander Pautsch
- Medicinal Chemistry , Boehringer Ingelheim Pharma GmbH & Co. KG , Birkendorfer Strasse 65 , 88397 Biberach an der Riss , Germany
| | - Alexander Weber
- Medicinal Chemistry , Boehringer Ingelheim Pharma GmbH & Co. KG , Birkendorfer Strasse 65 , 88397 Biberach an der Riss , Germany
| | - Christofer S Tautermann
- Medicinal Chemistry , Boehringer Ingelheim Pharma GmbH & Co. KG , Birkendorfer Strasse 65 , 88397 Biberach an der Riss , Germany
| |
Collapse
|
398
|
Charoenpattarapreeda J, Tan YS, Iegre J, Walsh SJ, Fowler E, Eapen RS, Wu Y, Sore HF, Verma CS, Itzhaki L, Spring DR. Targeted covalent inhibitors of MDM2 using electrophile-bearing stapled peptides. Chem Commun (Camb) 2019; 55:7914-7917. [PMID: 31225847 DOI: 10.1039/c9cc04022f] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Herein, we describe the development of a novel staple with an electrophilic warhead to enable the generation of stapled peptide covalent inhibitors of the p53-MDM2 protein-protein interaction (PPI). The peptide developed showed complete and selective covalent binding resulting in potent inhibition of p53-MDM2 PPI.
Collapse
Affiliation(s)
| | - Yaw Sing Tan
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore
| | - Jessica Iegre
- Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW, Cambridge, UK.
| | - Stephen J Walsh
- Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW, Cambridge, UK.
| | - Elaine Fowler
- Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW, Cambridge, UK.
| | - Rohan S Eapen
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Yuteng Wu
- Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW, Cambridge, UK.
| | - Hannah F Sore
- Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW, Cambridge, UK.
| | - Chandra S Verma
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore and Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore and School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 673551, Singapore
| | - Laura Itzhaki
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - David R Spring
- Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW, Cambridge, UK.
| |
Collapse
|
399
|
van den Bedem H, Wilson MA. Shining light on cysteine modification: connecting protein conformational dynamics to catalysis and regulation. JOURNAL OF SYNCHROTRON RADIATION 2019; 26:958-966. [PMID: 31274417 PMCID: PMC6613112 DOI: 10.1107/s160057751900568x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/25/2019] [Indexed: 05/04/2023]
Abstract
Cysteine is a rare but functionally important amino acid that is often subject to covalent modification. Cysteine oxidation plays an important role in many human disease processes, and basal levels of cysteine oxidation are required for proper cellular function. Because reactive cysteine residues are typically ionized to the thiolate anion (Cys-S-), their formation of a covalent bond alters the electrostatic and steric environment of the active site. X-ray-induced photo-oxidation to sulfenic acids (Cys-SOH) can recapitulate some aspects of the changes that occur under physiological conditions. Here we propose how site-specific cysteine photo-oxidation can be used to interrogate ensuing changes in protein structure and dynamics at atomic resolution. Although this powerful approach can connect cysteine covalent modification to global protein conformational changes and function, careful biochemical validation must accompany all such studies to exclude misleading artifacts. New types of X-ray crystallography experiments and powerful computational methods are creating new opportunities to connect conformational dynamics to catalysis for the large class of systems that use covalently modified cysteine residues for catalysis or regulation.
Collapse
Affiliation(s)
- Henry van den Bedem
- Bioscience Division, SLAC National Accelerator Laboratory, Stanford University, 2575 Sand Hill Road, Menlo Park, CA 94025, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
| | - Mark A Wilson
- Department of Biochemistry and the Redox Biology Center, University of Nebraska, Lincoln, NE 68588, USA
| |
Collapse
|
400
|
Li K, Kelly MA, Gao J. Biocompatible conjugation of Tris base to 2-acetyl and 2-formyl phenylboronic acid. Org Biomol Chem 2019; 17:5908-5912. [PMID: 31145403 PMCID: PMC6581600 DOI: 10.1039/c9ob00726a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We describe the biocompatible conjugation of the Tris base to 2-formyl and 2-acetylphenylboronic acid (abbreviated as 2-FPBA and 2-APBA respectively), which have emerged as a versatile chemotype for fast biocompatible conjugation reactions. Tris base was found to react with 2-FPBA/APBA to give oxazolidinoboronate (OzB) complexes, analogous to the thiazolidinoboronate (TzB) and imidazolidinoboronate (IzB) complex formation that we recently reported. The Tris conjugations proceed well in complex biological media, and in contrast to the TzB/IzB complexes, the Tris conjugates exhibit superior kinetic stability (dissociation over days) as well as chemical stability against oxidation. We demonstrate the utility of such conjugation chemistries via a small molecule-induced peptide cyclization in blood serum.
Collapse
Affiliation(s)
- Kaicheng Li
- Department of Chemistry, Merkert Chemistry Center, Boston College, 2609 Beacon Street, Chestnut Hill, Massachusetts 02467, USA.
| | - Michael A Kelly
- Department of Chemistry, Merkert Chemistry Center, Boston College, 2609 Beacon Street, Chestnut Hill, Massachusetts 02467, USA.
| | - Jianmin Gao
- Department of Chemistry, Merkert Chemistry Center, Boston College, 2609 Beacon Street, Chestnut Hill, Massachusetts 02467, USA.
| |
Collapse
|