1
|
Doherty K, Kessie K, Martin H, Loughlin J, Dulawa O, Kasements K, Velasco-Torrijos T. Synthesis of aromatic glycoconjugates as anti-fungal agents against Candida spp. and assessment of their covalent crosslinking capabilities. Bioorg Med Chem 2025; 117:118020. [PMID: 39612771 DOI: 10.1016/j.bmc.2024.118020] [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: 10/24/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024]
Abstract
Covalent drugs are becoming increasingly attractive in drug discovery, as they can enhance potency and selectivity for their molecular targets. Covalent inhibitors have been investigated for several therapeutic applications, including anti-cancer and anti-infection agents. However, there are only a few examples of covalent inhibitors targeting fungal pathogens. We have previously reported aromatic glycoconjugates (AGCs) capable of inhibiting the adhesion of Candida albicans to buccal epithelial cells. In this work, we synthesize novel derivatives of the AGCs to which we have added reactive functional groups, such as acryloyl and vinyl sulfones, and investigated their antifungal efficacy against Candida spp. Although the compounds were ineffective at clinically relevant concentrations, we found that some of the galactose derivatives featuring reactive groups were amongst the most active, so their ability to crosslink nucleophilic side chains was assessed in model reactions.
Collapse
Affiliation(s)
- Kyle Doherty
- Department of Chemistry, Maynooth University, Maynooth, Co. Kildare, Ireland; The Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Keela Kessie
- Department of Chemistry, Maynooth University, Maynooth, Co. Kildare, Ireland; The Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Harlei Martin
- Department of Chemistry, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Jordan Loughlin
- Department of Chemistry, Maynooth University, Maynooth, Co. Kildare, Ireland; The Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Oliwier Dulawa
- Department of Chemistry, Maynooth University, Maynooth, Co. Kildare, Ireland; The Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Kaja Kasements
- Laboratory of Environmental Toxicology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia.
| | - Trinidad Velasco-Torrijos
- Department of Chemistry, Maynooth University, Maynooth, Co. Kildare, Ireland; The Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland.
| |
Collapse
|
2
|
Cao Y, Yu T, Zhu Z, Zhang Y, Sun S, Li N, Gu C, Yang Y. Exploring the landscape of post-translational modification in drug discovery. Pharmacol Ther 2025; 265:108749. [PMID: 39557344 DOI: 10.1016/j.pharmthera.2024.108749] [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: 07/21/2024] [Revised: 09/11/2024] [Accepted: 11/04/2024] [Indexed: 11/20/2024]
Abstract
Post-translational modifications (PTMs) play a crucial role in regulating protein function, and their dysregulation is frequently associated with various diseases. The emergence of epigenetic drugs targeting factors such as histone deacetylases (HDACs) and histone methyltransferase enhancers of zeste homolog 2 (EZH2) has led to a significant shift towards precision medicine, offering new possibilities to overcome the limitations of traditional therapeutics. In this review, we aim to systematically explore how small molecules modulate PTMs. We discuss the direct targeting of enzymes involved in PTM pathways, the modulation of substrate proteins, and the disruption of protein-enzyme interactions that govern PTM processes. Additionally, we delve into the emerging strategy of employing multifunctional molecules to precisely regulate the modification levels of proteins of interest (POIs). Furthermore, we examine the specific characteristics of these molecules, evaluating their therapeutic benefits and potential drawbacks. The goal of this review is to provide a comprehensive understanding of PTM-targeting strategies and their potential for personalized medicine, offering a forward-looking perspective on the evolution of precision therapeutics.
Collapse
Affiliation(s)
- Yuhao Cao
- Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing 210022, China; School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tianyi Yu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ziang Zhu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuanjiao Zhang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shanliang Sun
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Nianguang Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Chunyan Gu
- Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing 210022, China; School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Ye Yang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
3
|
Tang T, Luo J, Zhang D, Lu Y, Liao W, Zhang J. Innovative design and potential applications of covalent strategy in drug discovery. Eur J Med Chem 2024; 284:117202. [PMID: 39756145 DOI: 10.1016/j.ejmech.2024.117202] [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: 08/29/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 01/07/2025]
Abstract
Covalent inhibitors provide persistent inhibition while maintaining excellent selectivity and efficacy by creating stable covalent connections with specific amino acids in target proteins. This technique enables the precise inhibition of previously undruggable targets, lowering the frequency of administration and potentially bypassing drug resistance. Because of these advantages, covalent inhibitors have tremendous potential in treating cancer, inflammation, and infectious illnesses, making them extremely important in modern pharmacological research. Covalent inhibitors targeting EGFR, BTK, and KRAS (G12X), which overcome drug resistance and off-target, non-"medicinal" difficulties, as well as covalent inhibitors targeting SARS-CoV-2 Mpro, have paved the way for the development of new antiviral medicines. Furthermore, the use of covalent methods in drug discovery procedures, such as covalent PROTACs, covalent molecular gels, covalent probes, CoLDR, and Dual-targeted covalent inhibitors, preserves these tactics' inherent features while incorporating the advantages of covalent inhibitors. This synthesis opens up new therapeutic opportunities. This review comprehensively examines the use of covalent techniques in drug discovery, emphasizing their transformational potential for future drug development.
Collapse
Affiliation(s)
- Tianyong Tang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxiang Luo
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Dan Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yang Lu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Wen Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Jifa Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
4
|
Zhao Z, Bourne PE. Exploring Extended Warheads toward Developing Cysteine-Targeted Covalent Kinase Inhibitors. J Chem Inf Model 2024; 64:9517-9527. [PMID: 39656065 DOI: 10.1021/acs.jcim.4c00890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
In designing covalent kinase inhibitors (CKIs), the inclusion of electrophiles as attacking warheads demands careful choreography, ensuring not only their presence on the scaffold moiety but also their precise interaction with nucleophiles in the binding sites. Given the limited number of known electrophiles, exploring adjacent chemical space to broaden the palette of available electrophiles capable of covalent inhibition is desirable. Here, we systematically analyze the characteristics of warheads and the corresponding adjacent fragments for use in CKI design. We first collect all the released cysteine-targeted CKIs from multiple databases and create one CKI data set containing 16,961 kinase-inhibitor data points from 12,381 unique CKIs covering 146 kinases with accessible cysteines in their binding pockets. Then, we analyze this data set, focusing on the extended warheads (i.e., warheads + adjacent fragments)─including 30 common warheads and 1344 unique adjacent fragments. In so doing, we provide structural insights and delineate chemical properties and patterns in these extended warheads. Notably, we highlight the popular patterns observed within reversible CKIs for the popular warheads cyanoacrylamide and aldehyde. This study provides medicinal chemists with novel insights into extended warheads and a comprehensive source of adjacent fragments, thus guiding the design, synthesis, and optimization of CKIs.
Collapse
Affiliation(s)
- Zheng Zhao
- School of Data Science and Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Philip E Bourne
- School of Data Science and Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22904, United States
| |
Collapse
|
5
|
Chen F, Liu Q, Ma L, Yan C, Zhang H, Zhou Z, Yi W. Identification of Novel Organo-Se BTSA-Based Derivatives as Potent, Reversible, and Selective PPARγ Covalent Modulators for Antidiabetic Drug Discovery. J Med Chem 2024. [PMID: 39705161 DOI: 10.1021/acs.jmedchem.4c02803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2024]
Abstract
Recent studies have identified selective peroxisome proliferator-activated receptor γ (PPARγ) modulators, which synergistically engage in the inhibition mechanism of PPARγ-Ser273 phosphorylation, as a promising approach for developing safer and more effective antidiabetic drugs. Herein, we present the design, synthesis, and evaluation of a new class of organo-Se compounds, namely, benzothiaselenazole-1-oxides (BTSAs), acting as potent, reversible, and selective PPARγ covalent modulators. Notably, 2n, especially (R)-2n, displayed a high binding affinity and superior antidiabetic effects with diminished side effects. This is mainly because it can reversibly form a unique covalent bond with the Cys285 residue in PPARγ-LBD. Further mechanistic investigations revealed that it manifested such desired pharmacological profiles primarily by effectively suppressing PPARγ-Ser273 phosphorylation, enhancing glucose metabolism, and selectively upregulating the expression of insulin-sensitive genes. Collectively, our results suggest that (R)-2n holds promise as a lead compound for treating T2DM and also provides an innovative reversible covalent warhead reference for future covalent drug design.
Collapse
Affiliation(s)
- Fangyuan Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Qingmei Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Lei Ma
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Cuishi Yan
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Haiman Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Zhi Zhou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Wei Yi
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| |
Collapse
|
6
|
Mathiesen IR, Calder EDD, Kunzelmann S, Walport LJ. Discovering covalent cyclic peptide inhibitors of peptidyl arginine deiminase 4 (PADI4) using mRNA-display with a genetically encoded electrophilic warhead. Commun Chem 2024; 7:304. [PMID: 39702664 DOI: 10.1038/s42004-024-01388-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/04/2024] [Indexed: 12/21/2024] Open
Abstract
Covalent drugs can achieve high potency with long dosing intervals. However, concerns remain about side-effects associated with off-target reactivity. Combining macrocyclic peptides with covalent warheads provides a solution to minimise off-target reactivity: the peptide enables highly specific target binding, positioning a weakly reactive warhead proximal to a suitable residue in the target. Here we demonstrate the direct discovery of covalent cyclic peptides using encoded libraries containing a weakly electrophilic cysteine-reactive fluoroamidine warhead. We combine direct incorporation of the warhead into peptide libraries using the flexible in vitro translation system with a peptide selection approach that identifies only covalent target binders. Using this approach, we identify potent and selective covalent inhibitors of the peptidyl arginine deiminase, PADI4 or PAD4, that react exclusively at the active site cysteine. We envisage this approach will enable covalent peptide inhibitor discovery for a range of related enzymes and expansion to alternative warheads in the future.
Collapse
Affiliation(s)
- Isabel R Mathiesen
- Protein-Protein Interaction Laboratory, The Francis Crick Institute, London, UK
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Ewen D D Calder
- Protein-Protein Interaction Laboratory, The Francis Crick Institute, London, UK
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Simone Kunzelmann
- Structural Biology Scientific Technology Platform, The Francis Crick Institute, London, UK
| | - Louise J Walport
- Protein-Protein Interaction Laboratory, The Francis Crick Institute, London, UK.
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK.
| |
Collapse
|
7
|
Jermusek FA, Webb LJ. Determining the Electrostatic Contributions of GTPase-GEF Complexes on Interfacial Drug Binding Specificity: A Case Study of a Protein-Drug-Protein Complex. Biochemistry 2024; 63:3220-3235. [PMID: 39589755 DOI: 10.1021/acs.biochem.4c00313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Understanding the factors that contribute to specificity of protein-protein interactions allows for design of orthosteric small molecules. Within this environment, a small molecule requires both structural and electrostatic complementarity. While the structural contribution to protein-drug-protein specificity is well characterized, electrostatic contributions require more study. To this end, we used a series of protein complexes involving Arf1 bound to guanine nucleotide exchange factors (GEFs) that are sensitive or resistant to the small molecule brefeldin A (BFA). By comparing BFA-sensitive Arf1-Gea1p and Arf1-ARNO with different combinations of four BFA sensitizing ARNO mutations (ARNOwt, ARNO1M, ARNO3M, and ARNO4M), we describe how electrostatic environments at each interface guide BFA binding specificity. We labeled Arf1 with cyanocysteine at several interfacial sites and measured by nitrile adsorption frequencies to map changes in electric field at each interface using the linear Stark equation. Temperature dependence of nitrile vibrational spectra was used to investigate differences in hydrogen bonding environments. These comparisons showed that interfacial electric field at the surface of Arf1 varied substantially depending on the GEF. The greatest differences were seen between Arf1-ARNOwt and Arf1-ARNO4M, suggesting a greater change in electric field is required for BFA binding to Arf1-ARNO. Additionally, rigidity of the interface of the Arf1-ARNO complex correlated strongly with BFA sensitivity, indicating that flexible interfaces are sensitive to disruption upon orthosteric small molecule binding. These findings demonstrate a qualitatively consistent electrostatic environment for Arf1 binding and more subtle differences preventing BFA specificity. We discuss how these results will guide improved design of other small molecules that can target protein-protein interfaces.
Collapse
Affiliation(s)
- Frank A Jermusek
- Interdisciplinary Life Sciences Graduate Program, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Lauren J Webb
- Interdisciplinary Life Sciences Graduate Program, The University of Texas at Austin, Austin, Texas 78712, United States
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
8
|
Klett T, Stahlecker J, Jaag S, Masberg B, Knappe C, Lämmerhofer M, Coles M, Stehle T, Boeckler FM. Covalent Fragments Acting as Tyrosine Mimics for Mutant p53-Y220C Rescue by Nucleophilic Aromatic Substitution. ACS Pharmacol Transl Sci 2024; 7:3984-3999. [PMID: 39698266 PMCID: PMC11651176 DOI: 10.1021/acsptsci.4c00414] [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: 08/06/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 12/20/2024]
Abstract
The tumor suppressor p53 is frequently mutated in human cancers. The Y220C mutant is the ninth most common p53 cancer mutant and is classified as a structural mutant, as it leads to strong thermal destabilization and degradation by creating a solvent-accessible hydrophobic cleft. To identify small molecules that thermally stabilize p53, we employed DSF to screen SNAr-type electrophiles from our covalent fragment library (CovLib) for binding to different structural (Y220C, R282W) and DNA contact (R273H) mutants of p53. The reactive fragments SN001, SN006, and SN007 were detected to specifically stabilize Y220C, indicating the arylation of Cys220 in the mutational cleft, as confirmed by X-ray crystallography. The fragments occupy the central cavity and mimic the ring system of the WT tyrosine lost by the mutation. Surpassing previously reported noncovalent ligands, SN001 stabilized T-p53C-Y220C concentration-dependently up to 4.45 °C and, due to its small size, represents a promising starting point for optimization.
Collapse
Affiliation(s)
- Theresa Klett
- Lab
for Molecular Design & Pharm. Biophysics, Institute of Pharmaceutical
Sciences, Eberhard Karls Universität
Tübingen, 72076 Tübingen, Germany
| | - Jason Stahlecker
- Lab
for Molecular Design & Pharm. Biophysics, Institute of Pharmaceutical
Sciences, Eberhard Karls Universität
Tübingen, 72076 Tübingen, Germany
| | - Simon Jaag
- Pharmaceutical
(Bio-)Analysis, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, 72076 Tübingen, Germany
| | - Benedikt Masberg
- Pharmaceutical
(Bio-)Analysis, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, 72076 Tübingen, Germany
| | - Cornelius Knappe
- Pharmaceutical
(Bio-)Analysis, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, 72076 Tübingen, Germany
| | - Michael Lämmerhofer
- Pharmaceutical
(Bio-)Analysis, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, 72076 Tübingen, Germany
| | - Murray Coles
- Department
of Protein Evolution, Max-Planck-Institute
for Biology, 72076 Tübingen, Germany
| | - Thilo Stehle
- Interfaculty
Institute of Biochemistry, Eberhard Karls
Universität Tübingen, 72076 Tübingen, Germany
| | - Frank M. Boeckler
- Lab
for Molecular Design & Pharm. Biophysics, Institute of Pharmaceutical
Sciences, Eberhard Karls Universität
Tübingen, 72076 Tübingen, Germany
- Interfaculty
Institute for Biomedical Informatics (IBMI), Eberhard Karls Universität Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
9
|
Wittlinger F, Chitnis SP, Pham CD, Damghani T, Patel KB, Möllers M, Schaeffner IK, Abidakun OA, Deng MQ, Ogboo BC, Rasch A, Beyett TS, Buckley B, Feru F, Shaurova T, Knappe C, Eck MJ, Hershberger PA, Scott DA, Brandt AL, Laufer SA, Heppner DE. Tilting the Scales toward EGFR Mutant Selectivity: Expanding the Scope of Bivalent "Type V" Kinase Inhibitors. J Med Chem 2024; 67:21438-21469. [PMID: 39626019 DOI: 10.1021/acs.jmedchem.4c02311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Binding multiple sites within proteins with bivalent compounds is a strategy for developing uniquely active agents. A new class of dual-site inhibitors has emerged targeting the epidermal growth factor receptor (EGFR) anchored to both the orthosteric (ATP) and allosteric sites. Despite proof-of-concept successes, enabling selectivity against oncogenic activating mutations has not been achieved and classifying these inhibitors among kinase inhibitors remains underexplored. This study investigates the structure-activity relationships, binding modes, and biological activity of ATP-allosteric bivalent inhibitors (AABIs). We find that AABIs selectively inhibit drug-resistant EGFR mutants (L858R/T790M and L858R/T790M/C797S) by anchoring a methyl isoindolinone moiety along the αC-helix channel of the allosteric site. In contrast, related Type I1/2 inhibitors target wild-type EGFR but are less effective against resistant mutants. This shift in selectivity demonstrates that mutant-selective AABIs classify as "Type V" bivalent inhibitors.
Collapse
Affiliation(s)
- Florian Wittlinger
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Surbhi P Chitnis
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Calvin D Pham
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Tahereh Damghani
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Kishan B Patel
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Mareike Möllers
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Ilse K Schaeffner
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Omobolanle A Abidakun
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Matthew Q Deng
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Blessing C Ogboo
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Alexander Rasch
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Tyler S Beyett
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Brian Buckley
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, United States
| | - Frederic Feru
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Tatiana Shaurova
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, United States
| | - Cornelius Knappe
- Institute of Pharmaceutical Sciences, Pharmaceutical (Bio-)Analysis, University of Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Michael J Eck
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Pamela A Hershberger
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, United States
| | - David A Scott
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Asher L Brandt
- Department of Chemistry, University of Saint Joseph, West Hartford, Connecticut 06117 United States
| | - Stefan A Laufer
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies" Eberhard Karls Universität Tübingen, 72076 Tübingen, Germany
- Tübingen Center for Academic Drug Discovery & Development (TüCAD2), 72076 Tübingen, Germany
| | - David E Heppner
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14203, United States
- Department of Structural Biology, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| |
Collapse
|
10
|
Zheng M, Kong L, Gao J. Boron enabled bioconjugation chemistries. Chem Soc Rev 2024; 53:11888-11907. [PMID: 39479937 PMCID: PMC11525960 DOI: 10.1039/d4cs00750f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Indexed: 11/02/2024]
Abstract
Novel bioconjugation reactions have been heavily pursued for the past two decades. A myriad of conjugation reactions have been developed for labeling molecules of interest in their native context as well as for constructing multifunctional molecular entities or stimuli-responsive materials. A growing cluster of bioconjugation reactions were realized by tapping into the unique properties of boron. As a rare element in human biology, boronic acids and esters exhibit remarkable biocompatibility. A number of organoboron reagents have been evaluated for bioconjugation, targeting the reactivity of either native biomolecules or those incorporating bioorthogonal functional groups. Owing to the dynamic nature of B-O and B-N bond formation, a significant portion of the boron-enabled bioconjugations exhibit rapid reversibility and accordingly have found applications in the development of reversible covalent inhibitors. On the other hand, stable bioconjugations have been developed that display fast kinetics and significantly expand the repertoire of bioorthogonal chemistry. This contribution presents a summary and comparative analysis of the recently developed boron-mediated bioconjugations. Importantly, this article seeks to provide an in-depth discussion of the thermodynamic and kinetic profiles of these boron-enabled bioconjugations, which reveals structure-reactivity relationships and provides guidelines for bioapplications.
Collapse
Affiliation(s)
- Mengmeng Zheng
- Department of Chemistry, Merkert Chemistry Center, Boston College, 2609 Beacon Street, Chestnut Hill, MA 02467, USA.
| | - Lingchao Kong
- Department of Chemistry, Merkert Chemistry Center, Boston College, 2609 Beacon Street, Chestnut Hill, MA 02467, USA.
| | - Jianmin Gao
- Department of Chemistry, Merkert Chemistry Center, Boston College, 2609 Beacon Street, Chestnut Hill, MA 02467, USA.
| |
Collapse
|
11
|
Zhang S, Wang L, Lu Y, Guo C, Zhang T, Zhang L. Targeting spleen tyrosine kinase (SYK): structure, mechanisms and drug discovery. Drug Discov Today 2024; 30:104257. [PMID: 39653169 DOI: 10.1016/j.drudis.2024.104257] [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: 07/01/2024] [Revised: 11/23/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024]
Abstract
Spleen tyrosine kinase (SYK) is a crucial non-receptor tyrosine kinase involved in signaling pathways that regulate various cellular processes. It is primarily expressed in hematopoietic cells and myeloid cells, which are crucial for B-cell development, maturation and antibody production, and it is a key therapeutic target for autoimmune and allergic diseases. Overexpression of SYK is also associated with cancer and cardiovascular, cerebrovascular and neurodegenerative diseases, contributing to their initiation and progression. SYK is a promising target for drug development, and several inhibitors have already been reported. This review covers the structure and regulatory pathways of SYK, as well as its links to various diseases. It also highlights key small-molecule SYK inhibitors, their design strategies and their potential therapeutic benefits, aiming to enhance our understanding and aid in the discovery of more-effective SYK inhibitors.
Collapse
Affiliation(s)
- Shuangqian Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Lilin Wang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China; Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China
| | - Yingying Lu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Chuanxin Guo
- Nucleic Acid Division, Shanghai Cell Therapy Group, Shanghai 201805, China.
| | - Tongtong Zhang
- Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China; The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China.
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| |
Collapse
|
12
|
Torres-Gómez H, Keiff F, Hortschansky P, Bernal F, Kerndl V, Meyer F, Messerschmidt N, Dal Molin M, Krüger T, Rybniker J, Brakhage AA, Kloss F. Replacement of the essential nitro group by electrophilic warheads towards nitro-free antimycobacterial benzothiazinones. Eur J Med Chem 2024; 279:116849. [PMID: 39265253 DOI: 10.1016/j.ejmech.2024.116849] [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: 06/25/2024] [Revised: 08/24/2024] [Accepted: 09/03/2024] [Indexed: 09/14/2024]
Abstract
Nitrobenzothiazinones (BTZs) are undergoing late-stage development as a novel class of potent antitubercular drug candidates with two compounds in clinical phases. BTZs inhibit decaprenylphosphoryl-β-d-ribose oxidase 1 (DprE1), a key enzyme in cell wall biosynthesis of mycobacteria. Their mechanism of action involves an in-situ-reduction of the nitro moiety to a reactive nitroso intermediate capable of covalent binding to Cys387 in the catalytic cavity. The electron-deficient nature of the aromatic core is a key driver for the formation of hydride-Meisenheimer complexes (HMC) as main metabolites in vivo. To mimic the electrophilic character of the nitroso moiety, bioisosteric replacement with different electrophilic warheads was attempted to reduce HMC formation without compromising covalent reactivity. Herein, we synthesized and characterized various covalent warheads covering different reaction principles. Covalent inhibition was confirmed for most active antimycobacterial compounds by enzymatic inhibition assays and peptide fragment analysis.
Collapse
Affiliation(s)
- Héctor Torres-Gómez
- Transfer Group Anti-infectives, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), Adolf-Reichwein-Str. 23, 07745, Jena, Germany
| | - François Keiff
- Transfer Group Anti-infectives, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), Adolf-Reichwein-Str. 23, 07745, Jena, Germany
| | - Peter Hortschansky
- Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), Adolf-Reichwein- Str. 23, 07745, Jena, Germany
| | - Freddy Bernal
- Transfer Group Anti-infectives, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), Adolf-Reichwein-Str. 23, 07745, Jena, Germany
| | - Valerie Kerndl
- Transfer Group Anti-infectives, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), Adolf-Reichwein-Str. 23, 07745, Jena, Germany
| | - Florian Meyer
- Transfer Group Anti-infectives, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), Adolf-Reichwein-Str. 23, 07745, Jena, Germany
| | - Nina Messerschmidt
- Transfer Group Anti-infectives, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), Adolf-Reichwein-Str. 23, 07745, Jena, Germany
| | - Michael Dal Molin
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Thomas Krüger
- Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), Adolf-Reichwein- Str. 23, 07745, Jena, Germany
| | - Jan Rybniker
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany; German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 50931, Cologne, Germany
| | - Axel A Brakhage
- Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), Adolf-Reichwein- Str. 23, 07745, Jena, Germany; Institute of Microbiology, Friedrich Schiller University, Adolf-Reichwein-Str. 23, 07745, Jena, Germany
| | - Florian Kloss
- Transfer Group Anti-infectives, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), Adolf-Reichwein-Str. 23, 07745, Jena, Germany.
| |
Collapse
|
13
|
Li Y, Zhang Y, Zhang J, Zhan Z, Mao W. Development of novel focal adhesion kinase (FAK) inhibitors for targeting cancer: Structural insights and therapeutic potential. Eur J Med Chem 2024; 279:116913. [PMID: 39357313 DOI: 10.1016/j.ejmech.2024.116913] [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: 08/23/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024]
Abstract
Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase frequently overexpressed in various cancer cells, facilitating tumor growth through the regulation of cell adhesion, migration, and proliferation. Consequently, targeting FAK is considered a promising anti-tumor strategy, particularly for invasive cancers. Numerous potent small-molecule inhibitors have progressed to clinical trials. Among these, Defactinib is under evaluation for regulatory approval as a treatment for ovarian serous tumors. Furthermore, novel FAK inhibitors, including PROTACs, have emerged as key research focuses, anticipated to overcome the limitations of traditional inhibitors. In this Perspective, we highlight the protein structure, biological functions, relevant signaling pathways, and associations of FAK with cancer development. We also analyze the clinical status of FAK inhibitors, paying special attention to the various classes of FAK inhibitors, with detailed analyses of their chemical structures, structure-activity relationships (SARs), bioactivity profiles, selectivity profiles, and therapeutic potentials.
Collapse
Affiliation(s)
- Yingnan Li
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Neuro-system and Multimorbidity Laboratory, State Key Laboratory of Biotherapy and Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610041, Sichuan, China
| | - Yuming Zhang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Neuro-system and Multimorbidity Laboratory, State Key Laboratory of Biotherapy and Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610041, Sichuan, China; West China College of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jifa Zhang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Neuro-system and Multimorbidity Laboratory, State Key Laboratory of Biotherapy and Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610041, Sichuan, China
| | - Zixuan Zhan
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Neuro-system and Multimorbidity Laboratory, State Key Laboratory of Biotherapy and Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610041, Sichuan, China.
| | - Wuyu Mao
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Neuro-system and Multimorbidity Laboratory, State Key Laboratory of Biotherapy and Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610041, Sichuan, China.
| |
Collapse
|
14
|
Wilders H, Biggs G, Rowe SM, Cawood EE, Riziotis IG, Rendina AR, Grant EK, Pettinger J, Fallon DJ, Skehel M, House D, Tomkinson NCO, Bush JT. Expedited SARS-CoV-2 Main Protease Inhibitor Discovery through Modular 'Direct-to-Biology' Screening. Angew Chem Int Ed Engl 2024:e202418314. [PMID: 39630105 DOI: 10.1002/anie.202418314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/15/2024] [Indexed: 12/12/2024]
Abstract
Reactive fragment (RF) screening has emerged as an efficient method for ligand discovery across the proteome, irrespective of a target's perceived tractability. To date, however, the efficiency of subsequent optimisation campaigns has largely been low-throughput, constrained by the need for synthesis and purification of target compounds. We report an efficient platform for 'direct-to-biology' (D2B) screening of cysteine-targeting chloroacetamide RFs, wherein synthesis is performed in 384-well plates allowing direct assessment in downstream biological assays without purification. Here, the developed platform was used to optimise inhibitors of SARS-CoV-2 main protease (MPro), an established drug target for the treatment of COVID-19. An initial RF hit was developed into a series of potent inhibitors, and further exploration using D2B screening enabled a 'switch' to a reversible inhibitor series. This example of ligand discovery for MPro illustrates the acceleration that D2B chemistry can offer for optimising RFs towards covalent inhibitor candidates, as well as providing future impetus to explore the evolution of RFs into non-covalent ligands.
Collapse
Affiliation(s)
- Harry Wilders
- Chemical Biology, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
- Crick-GSK Biomedical Linklabs, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
- Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow, G1 1XL, UK
| | - George Biggs
- Crick-GSK Biomedical Linklabs, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Sam M Rowe
- Chemical Biology, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Emma E Cawood
- Crick-GSK Biomedical Linklabs, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Ioannis G Riziotis
- Crick-GSK Biomedical Linklabs, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Alan R Rendina
- Screening, Profiling and Mechanistic Biology, GSK, 1250 South Collegeville Road, Collegevill, PA, 19426, US
| | - Emma K Grant
- Chemical Biology, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Jonathan Pettinger
- Chemical Biology, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
- Crick-GSK Biomedical Linklabs, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - David J Fallon
- Chemical Biology, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Mark Skehel
- Proteomics Science Technology Platform, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - David House
- Chemical Biology, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
- Crick-GSK Biomedical Linklabs, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Nicholas C O Tomkinson
- Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow, G1 1XL, UK
| | - Jacob T Bush
- Chemical Biology, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
- Crick-GSK Biomedical Linklabs, GSK, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
- Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow, G1 1XL, UK
| |
Collapse
|
15
|
Liang J, Huang J, Yang J, Liang W, Li H, Wu Y, Liu B. Synthesis and in vitro evaluation of benzo[b]thiophene-3-carboxylic acid 1,1-dioxide derivatives as anticancer agents targeting the RhoA/ROCK pathway. J Enzyme Inhib Med Chem 2024; 39:2390911. [PMID: 39258708 PMCID: PMC11391881 DOI: 10.1080/14756366.2024.2390911] [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: 04/27/2024] [Revised: 06/24/2024] [Accepted: 07/18/2024] [Indexed: 09/12/2024] Open
Abstract
Rho family GTPases regulate cellular processes and promote tumour growth and metastasis; thus, RhoA is a potential target for tumour metastasis inhibition. However, limited progress has been made in the development of RhoA targeting anticancer drugs. Here, we synthesised benzo[b]thiophene-3-carboxylic acid 1,1-dioxide derivatives based on a covalent inhibitor of RhoA (DC-Rhoin), reported in our previous studies. The observed structure-activity relationship (contributed by carboxamide in C-3 and 1-methyl-1H-pyrazol in C-5) enhanced the anti-proliferative activity of the derivatives. Compound b19 significantly inhibited the proliferation, migration, and invasion of MDA-MB-231 cells and promoted their apoptosis. The suppression of myosin light chain phosphorylation and the formation of stress fibres confirmed the inhibitory activity of b19 via the RhoA/ROCK pathway. b19 exhibited a different binding pattern from DC-Rhoin, as observed in molecular docking analysis. This study provides a reference for the development of anticancer agents targeting the RhoA/ROCK pathway.
Collapse
Affiliation(s)
- Jinhao Liang
- The Second Clinical Medical College, and Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jin Huang
- The Second Clinical Medical College, and Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianzhan Yang
- The Second Clinical Medical College, and Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weihong Liang
- The Second Clinical Medical College, and Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangzhou Key Laboratory of Chirality Research on Active Components of Traditional Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haoxiang Li
- The Second Clinical Medical College, and Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yunshan Wu
- The Second Clinical Medical College, and Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangzhou Key Laboratory of Chirality Research on Active Components of Traditional Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bo Liu
- The Second Clinical Medical College, and Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangzhou Key Laboratory of Chirality Research on Active Components of Traditional Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
16
|
Shang J, Yin S, Shen J, Gao C, Wang W, Sun K, Zhu W, Fu P. Amino-Based Probe for Natural Products with Covalent Binding Ability to Lysine and Mechanism of Action of Medermycin. Org Lett 2024; 26:10129-10134. [PMID: 39560365 DOI: 10.1021/acs.orglett.4c03803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
A chemoselective amino-based probe was designed for discovering natural products with covalent binding potential to lysine. Using this reactivity-based technique, a marine-derived Streptomyces strain was identified, which could produce medermycin as the major metabolite. A new compound, mederpyrrole A, derived from medermycin and anthranilic acid through nonenzymatic reaction was isolated. Medermycin can react with primary amines under mild conditions to generate chimeric products possessing a naphthoquinone-pyrrole skeleton. It can also covalently bind to proteins.
Collapse
Affiliation(s)
- Jiaxu Shang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Shupeng Yin
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Jingjing Shen
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Chao Gao
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Weihong Wang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Ke Sun
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Weiming Zhu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Peng Fu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| |
Collapse
|
17
|
Cao H, Huang Z, Liu Z, Zhang X, Ren Y, Hameed MS, Rao L, Makunga NP, Dobrikov GM, Wan J. Structure-Guided Design of Affinity/Covalent-Bond Dual-Driven Inhibitors Targeting the AMP Site of FBPase. J Med Chem 2024; 67:20421-20437. [PMID: 39520680 DOI: 10.1021/acs.jmedchem.4c01886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Fructose-1,6-bisphosphatase (FBPase) has attracted substantial interest as a target associated with cancer and type II diabetes. FBPase inhibitors targeting the AMP allosteric site have been documented, but their limited selectivity has raised concerns about adverse effects. To address this issue, we designed the affinity/covalent-bond dual-driven inhibitors based on the pharmacophore knowledge of the AMP pocket and neighboring cysteine residue (C179) of FBPase using the cysteine-targeting reactivity warhead screen followed by a structural optimization strategy. Pull-down and Western Blotting assays confirmed FBPase as a direct target in hepatic cells. X-ray cocrystallographic structure of FBPase-11 and Cov_DOX calculation demonstrated that hydrogen bonding and π-π stacking were the predominant driving force for the inhibition of sulfonylurea-based FBPase covalent inhibitors, while covalent binding with C179 enhances the inhibitors' long-lasting hypoglycemic effects. Together, this work highlights the potential of affinity/covalent-bond dual-driven inhibitors in drug development and provides a promising approach for developing potent drugs targeting AMP-associated proteins.
Collapse
Affiliation(s)
- Hongxuan Cao
- State Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Zeyue Huang
- State Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Zheng Liu
- State Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Xiao Zhang
- State Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Yanliang Ren
- State Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Muhammad Salman Hameed
- State Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Li Rao
- State Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Nokwanda P Makunga
- Department of Botany and Zoology, Stellenbosch University, Private Bag X1, Matieland, Stellenbosch 7602, South Africa
| | - Georgi M Dobrikov
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Street, Bl. 9, 1113 Sofia, Bulgaria
| | - Jian Wan
- State Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China
| |
Collapse
|
18
|
Alboreggia G, Udompholkul P, Atienza EL, Muzzarelli K, Assar Z, Pellecchia M. Covalent Targeting of Histidine Residues with Aryl Fluorosulfates: Application to Mcl-1 BH3 Mimetics. J Med Chem 2024; 67:20214-20223. [PMID: 39532346 PMCID: PMC11613628 DOI: 10.1021/acs.jmedchem.4c01541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/08/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Covalent drugs provide pharmacodynamic and pharmacokinetic advantages over reversible agents. However, covalent strategies have been developed mostly to target cysteine (Cys) residues, which are rarely found in binding sites. Among other nucleophilic residues that could be in principle used for the design of covalent drugs, histidine (His) has not been given proper attention despite being in principle an attractive residue to pursue but underexplored. Aryl fluorosulfates, a mild electrophile that is very stable in biological media, have been recently identified as possible electrophiles to react with the side chains of Lys; however, limited studies are available on aryl fluorosulfates' ability to target His residues. We demonstrate that proper incorporation of an aryl fluorosulfate juxtaposing the electrophile with a His residue can be used to afford rapid optimizations of His-covalent agents. As an application, we report on His-covalent BH3 mimetics targeting His224 of Mcl-1.
Collapse
Affiliation(s)
- Giulia Alboreggia
- Division
of Biomedical Sciences, School of Medicine, University of California Riverside, 900 University Avenue, Riverside, California 92521, United States
| | - Parima Udompholkul
- Division
of Biomedical Sciences, School of Medicine, University of California Riverside, 900 University Avenue, Riverside, California 92521, United States
| | - Emma L. Atienza
- Division
of Biomedical Sciences, School of Medicine, University of California Riverside, 900 University Avenue, Riverside, California 92521, United States
| | - Kendall Muzzarelli
- Cayman Chemical
Co., 1180 E. Ellsworth Road, Ann Arbor, Michigan 48108, United States
| | - Zahra Assar
- Cayman Chemical
Co., 1180 E. Ellsworth Road, Ann Arbor, Michigan 48108, United States
| | - Maurizio Pellecchia
- Division
of Biomedical Sciences, School of Medicine, University of California Riverside, 900 University Avenue, Riverside, California 92521, United States
| |
Collapse
|
19
|
Liu C, Zhang M, Zeng L, Wan Y, Dai M. Ten-Step Total Synthesis of (±)-Phaeocaulisin A Enabled by Cyclopropanol Ring-Opening Carbonylation. J Am Chem Soc 2024; 146:32276-32282. [PMID: 39532298 PMCID: PMC11613322 DOI: 10.1021/jacs.4c12121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
We report an efficient total synthesis of (±)-phaeocaulisin A, a guaianolide sesquiterpene natural product possessing a complex tetracyclic skeleton embedded with an oxaspirolactone and a fused bicyclic lactone, four oxygen-containing stereocenters, and an 8-oxabicyclo[3.2.1]octane core. Our synthesis features a novel palladium-catalyzed cyclopropanol ring-opening carbonylation to access a key γ-ketoester, a chemo- and stereoselective aldol cyclization to form the seven-membered carbocycle, and a cascade ketalization-lactonization to construct the desired tetracyclic skeleton. With these strategically important C-C and C-O bond formation transformations, a 10-step total synthesis of (±)-phaeocaulisin A was achieved. We further developed the cyclopropanol ring-opening carbonylation chemistry to provide an alternative approach to prepare γ-ketoesters. Biologically, the penultimate intermediate with an α-methylene γ-butyrolactone moiety was identified as a promising lead compound with anticancer proliferation activity against a panel of triple-negative or HER2+ breast cancer cell lines.
Collapse
Affiliation(s)
- Chang Liu
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Mingyu Zhang
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Lidan Zeng
- Department
of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, Georgia 30322, United States
| | - Yong Wan
- Department
of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, Georgia 30322, United States
| | - Mingji Dai
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
- Department
of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
20
|
Raouf YS, Moreno-Yruela C. Slow-Binding and Covalent HDAC Inhibition: A New Paradigm? JACS AU 2024; 4:4148-4161. [PMID: 39610753 PMCID: PMC11600154 DOI: 10.1021/jacsau.4c00828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/30/2024]
Abstract
The dysregulated post-translational modification of proteins is an established hallmark of human disease. Through Zn2+-dependent hydrolysis of acyl-lysine modifications, histone deacetylases (HDACs) are key regulators of disease-implicated signaling pathways and tractable drug targets in the clinic. Early targeting of this family of 11 enzymes (HDAC1-11) afforded a first generation of broadly acting inhibitors with medicinal applications in oncology, specifically in cutaneous and peripheral T-cell lymphomas and in multiple myeloma. However, first-generation HDAC inhibitors are often associated with weak-to-modest patient benefits, dose-limited efficacies, pharmacokinetic liabilities, and recurring clinical toxicities. Alternative inhibitor design to target single enzymes and avoid toxic Zn2+-binding moieties have not overcome these limitations. Instead, recent literature has seen a shift toward noncanonical mechanistic approaches focused on slow-binding and covalent inhibition. Such compounds hold the potential of improving the pharmacokinetic and pharmacodynamic profiles of HDAC inhibitors through the extension of the drug-target residence time. This perspective aims to capture this emerging paradigm and discuss its potential to improve the preclinical/clinical outlook of HDAC inhibitors in the coming years.
Collapse
Affiliation(s)
- Yasir S. Raouf
- Department
of Chemistry, United Arab Emirates University, P.O. Box No. 15551 Al Ain, UAE
| | - Carlos Moreno-Yruela
- Laboratory
of Chemistry and Biophysics of Macromolecules (LCBM), Institute of
Chemical Sciences and Engineering (ISIC), School of Basic Sciences, École Polytechnique Fédérale
de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
21
|
Klahn P. How Should we Teach Medicinal Chemistry in Higher Education to Prepare Students for a Future Career as Medicinal Chemists and Drug Designers? - A Teacher's Perspective. ChemMedChem 2024:e202400791. [PMID: 39564941 DOI: 10.1002/cmdc.202400791] [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: 10/04/2024] [Indexed: 11/21/2024]
Abstract
In the recent two decades, the multidisciplinary field of medicinal chemistry has undergone several conceptual and technology-driven paradigm changes with significant impact on the skill set medicinal chemists need to acquire during their education. Considering the need for academic medicinal chemistry teaching, this article aims at identifying important skills, competences, and basic knowledge as general learning outcomes based on an analysis of the relevant stakeholders and concludes effective teaching strategies preparing students for a future career as medicinal chemists and drug designers.
Collapse
Affiliation(s)
- Philipp Klahn
- Department of Chemistry and Molecular Biology, Division of Organic and Medicinal Chemistry, University of Gothenburg, Medicinaregatan 7B, Natrium, Göteborg, 413 90, Sweden
| |
Collapse
|
22
|
Shen W, Chen X, Zhou L, Cheng Y, Zhang Y, Jiang X, Sun H, Shen J. Discovery of the potent covalent inhibitor with an acrylate warhead for SARS-CoV-2 3CL protease. Bioorg Med Chem Lett 2024; 112:129942. [PMID: 39218405 DOI: 10.1016/j.bmcl.2024.129942] [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: 04/21/2024] [Revised: 08/21/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
COVID-19 has caused severe consequences in terms of public health and economy worldwide since its outbreak in December 2019. SARS-CoV-2 3C-like protease (3CLpro), crucial for the viral replications, is an attractive target for the development of antiviral drugs. In this study, several kinds of Michael acceptor warheads were utilized to hunt for potent covalent inhibitors against 3CLpro. Meanwhile, novel 3CLpro inhibitors with the P3-3,5-dichloro-4-(2-(dimethylamino)ethoxy)phenyl moiety were designed and synthesized which may form salt bridge with residue Glu166. Among them, two compounds 12b and 12c exhibited high inhibitory activities against SARS-CoV-2 3CLpro. Further investigations suggested that 12b with an acrylate warhead displayed potent activity against HCoV-OC43 (EC50 = 97 nM) and SARS-CoV-2 replicon (EC50 = 45 nM) and low cytotoxicity (CC50 > 10 μM) in Huh7 cells. Taken together, this study devised two series of 3CLpro inhibitors and provided the potent SARS-CoV-2 3CLpro inhibitor (12b) which may be used for treating coronavirus infections.
Collapse
Affiliation(s)
- Wen Shen
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xinyao Chen
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; Yangtze Delta Drug Advanced Research Institute and Yangtze Delta Pharmaceutical College, Nantong 226133, China
| | - Liping Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Cheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiangrui Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haiguo Sun
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Jingshan Shen
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
23
|
Takasaki R, Ito E, Nagae M, Takahashi Y, Matsuoka T, Yasue W, Arichi N, Ohno H, Yamasaki S, Inuki S. Development of Ribityllumazine Analogue as Mucosal-Associated Invariant T Cell Ligands. J Am Chem Soc 2024; 146:29964-29976. [PMID: 39432319 DOI: 10.1021/jacs.4c12997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Mucosal-associated invariant T (MAIT) cells are a subset of innate-like T cells abundant in human tissues that play a significant role in defense against bacterial and viral infections and in tissue repair. MAIT cells are activated by recognizing microbial-derived small-molecule ligands presented by the MHC class I related-1 protein. Although several MAIT cell modulators have been identified in the past decade, potent and chemically stable ligands remain limited. Herein, we carried out a structure-activity relationship study of ribityllumazine derivatives and found a chemically stable MAIT cell ligand with a pteridine core and a 2-oxopropyl group as the Lys-reactive group. The ligand showed high potency in a cocultivation assay using model cell lines of antigen-presenting cells and MAIT cells. The X-ray crystallographic analysis revealed the binding mode of the ligand to MR1 and the T cell receptor, indicating that it forms a covalent bond with MR1 via Schiff base formation. Furthermore, we found that the ligand stimulated proliferation of human MAIT cells in human peripheral blood mononuclear cells and showed an adjuvant effect in mice. Our developed ligand is one of the most potent among chemically stable MAIT cell ligands, contributing to accelerating therapeutic applications of MAIT cells.
Collapse
Affiliation(s)
- Ryosuke Takasaki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Kyoto 606-8501, Japan
| | - Emi Ito
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka 565-0871, Japan
| | - Masamichi Nagae
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka 565-0871, Japan
| | - Yuki Takahashi
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Kyoto 606-8501, Japan
| | - Takuro Matsuoka
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Kyoto 606-8501, Japan
| | - Wakana Yasue
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Kyoto 606-8501, Japan
| | - Norihito Arichi
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Kyoto 606-8501, Japan
| | - Hiroaki Ohno
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Kyoto 606-8501, Japan
| | - Sho Yamasaki
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka 565-0871, Japan
| | - Shinsuke Inuki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Kyoto 606-8501, Japan
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Tokushima 770-8505, Japan
| |
Collapse
|
24
|
Du H, Zhang X, Wu Z, Zhang O, Gu S, Wang M, Zhu F, Li D, Hou T, Pan P. CovalentInDB 2.0: an updated comprehensive database for structure-based and ligand-based covalent inhibitor design and screening. Nucleic Acids Res 2024:gkae946. [PMID: 39441070 DOI: 10.1093/nar/gkae946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/23/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024] Open
Abstract
The rational design of targeted covalent inhibitors (TCIs) has emerged as a powerful strategy in drug discovery, known for its ability to achieve strong binding affinity and prolonged target engagement. However, the development of covalent drugs is often challenged by the need to optimize both covalent warhead and non-covalent interactions, alongside the limitations of existing compound libraries. To address these challenges, we present CovalentInDB 2.0, an updated online database designed to support covalent drug discovery. This updated version includes 8303 inhibitors and 368 targets, supplemented by 3445 newly added cocrystal structures, providing detailed analyses of non-covalent interactions. Furthermore, we have employed an AI-based model to profile the ligandability of 144 864 cysteines across the human proteome. CovalentInDB 2.0 also features the largest covalent virtual screening library with 2 030 192 commercially available compounds and a natural product library with 105 901 molecules, crucial for covalent drug screening and discovery. To enhance the utility of these compounds, we performed structural similarity analysis and drug-likeness predictions. Additionally, a new user data upload feature enables efficient data contribution and continuous updates. CovalentInDB 2.0 is freely accessible at http://cadd.zju.edu.cn/cidb/.
Collapse
Affiliation(s)
- Hongyan Du
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Xujun Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Zhenxing Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Odin Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Shukai Gu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Mingyang Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Dan Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Tingjun Hou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Peichen Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| |
Collapse
|
25
|
Fu S, Chen Z, Luo Z, Nie M, Fu T, Zhou Y, Yang Q, Zhu F, Ni F. Chem(Pro)2: the atlas of chemoproteomic probes labelling human proteins. Nucleic Acids Res 2024:gkae943. [PMID: 39436046 DOI: 10.1093/nar/gkae943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/25/2024] [Accepted: 10/11/2024] [Indexed: 10/23/2024] Open
Abstract
Chemoproteomic probes (CPPs) have been widely considered as powerful molecular biological tools that enable the highly efficient discovery of both binding proteins and modes of action for the studied compounds. They have been successfully used to validate targets and identify binders. The design of CPP has been considered extremely challenging, which asks for the generalization using a large number of probe data. However, none of the existing databases gives such valuable data of CPPs. Herein, a database entitled 'Chem(Pro)2' was therefore developed to systematically describe the atlas of diverse types of CPPs labelling human protein in living cell/lysate. With the booming application of chemoproteomic technique and artificial intelligence in current chemical biology study, Chem(Pro)2 was expected to facilitate the AI-based learning of interacting pattern among molecules for discovering innovative targets and new drugs. Till now, Chem(Pro)2 has been open to all users without any login requirement at: https://idrblab.org/chemprosquare/.
Collapse
Affiliation(s)
- Songsen Fu
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
- LeadArt Biotechnologies Ltd., Ningbo 315201, China
| | - Zhen Chen
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Zhiming Luo
- LeadArt Biotechnologies Ltd., Ningbo 315201, China
| | - Meiyun Nie
- LeadArt Biotechnologies Ltd., Ningbo 315201, China
| | - Tingting Fu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Ying Zhou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Qingxia Yang
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Feng Ni
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
- LeadArt Biotechnologies Ltd., Ningbo 315201, China
| |
Collapse
|
26
|
Merten EM, Sears JD, Leisner TM, Hardy PB, Ghoshal A, Hossain MA, Asressu KH, Brown PJ, Tse EG, Stashko MA, Li K, Norris-Drouin JL, Herring LE, Mordant AL, Webb TS, Mills CA, Barker NK, Streblow ZJ, Perveen S, Arrowsmith CH, Couñago RM, Arnold JJ, Cameron CE, Streblow DN, Moorman NJ, Heise MT, Willson TM, Popov KI, Pearce KH. Identification of a cell-active chikungunya virus nsP2 protease inhibitor using a covalent fragment-based screening approach. Proc Natl Acad Sci U S A 2024; 121:e2409166121. [PMID: 39388272 PMCID: PMC11494320 DOI: 10.1073/pnas.2409166121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that has been responsible for numerous large-scale outbreaks in the last twenty years. Currently, there are no FDA-approved therapeutics for any alphavirus infection. CHIKV nonstructural protein 2 (nsP2), which contains a cysteine protease domain, is essential for viral replication, making it an attractive target for a drug discovery campaign. Here, we optimized a CHIKV nsP2 protease (nsP2pro) biochemical assay for the screening of a 6,120-compound cysteine-directed covalent fragment library. Using a 50% inhibition threshold, we identified 153 hits (2.5% hit rate). In dose-response follow-up, RA-0002034, a covalent fragment that contains a vinyl sulfone warhead, inhibited CHIKV nsP2pro with an IC50 of 58 ± 17 nM, and further analysis with time-dependent inhibition studies yielded a kinact /KI of 6.4 × 103 M-1s-1. LC-MS/MS analysis determined that RA-0002034 covalently modified the catalytic cysteine in a site-specific manner. Additionally, RA-0002034 showed no significant off-target reactivity in proteomic experiments or against a panel of cysteine proteases. In addition to the potent biochemical inhibition of CHIKV nsP2pro activity and exceptional selectivity, RA-0002034 was tested in cellular models of alphavirus infection and effectively inhibited viral replication of both CHIKV and related alphaviruses. This study highlights the identification and characterization of the chemical probe RA-0002034 as a promising hit compound from covalent fragment-based screening for development toward a CHIKV or pan-alphavirus therapeutic.
Collapse
Affiliation(s)
- Eric M. Merten
- Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - John D. Sears
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Tina M. Leisner
- Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - P. Brian Hardy
- Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Anirban Ghoshal
- Structural Genomics Consortium, Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Mohammad Anwar Hossain
- Structural Genomics Consortium, Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Kesatebrhan Haile Asressu
- Structural Genomics Consortium, Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Peter J. Brown
- Structural Genomics Consortium, Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Edwin G. Tse
- Structural Genomics Consortium, Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Michael A. Stashko
- Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Kelin Li
- Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Jacqueline L. Norris-Drouin
- Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Laura E. Herring
- Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Angie L. Mordant
- Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Thomas S. Webb
- Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Christine A. Mills
- Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Natalie K. Barker
- Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Zachary J. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR97006
| | - Sumera Perveen
- Structural Genomics Consortium, University of Toronto, Toronto, ONM5G 1L7, Canada
| | - Cheryl H. Arrowsmith
- Structural Genomics Consortium, University of Toronto, Toronto, ONM5G 1L7, Canada
| | - Rafael Miguez Couñago
- Structural Genomics Consortium, Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Center of Medicinal Chemistry, Center for Molecular Biology and Genetic Engineering, University of Campinas, Campinas, SP13083-886, Brazil
| | - Jamie J. Arnold
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Craig E. Cameron
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Daniel N. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR97006
| | - Nathaniel J. Moorman
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Mark T. Heise
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Timothy M. Willson
- Structural Genomics Consortium, Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Konstantin I. Popov
- Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Kenneth H. Pearce
- Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Lineberger Comprehensive Cancer Center, Molecular Therapeutics Research Program, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| |
Collapse
|
27
|
Jackson PA, Kisty E, Pradhan V, Swank C, Bohrer L, Nolan TL, Weerapana E, Lapinsky DJ. Appendage- and Scaffold-Diverse Electrophilic and Photoreactive Probes for Integrated Phenotypic Screening-Target Identification Campaigns via a Minimalist Bifunctional Isocyanide. ACS OMEGA 2024; 9:42557-42570. [PMID: 39431108 PMCID: PMC11483914 DOI: 10.1021/acsomega.4c06879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/16/2024] [Accepted: 09/20/2024] [Indexed: 10/22/2024]
Abstract
One of the grand challenges in chemical biology is identifying a small-molecule modulator for all proteins within a proteome. To expand the variety and number of ligandable proteins for drug discovery, the objective of this study was to synthesize and evaluate the protein target profiles of electrophilic and photoreactive fully functionalized small-molecule probes (FFSMPs) featuring increased scaffold-, appendage-, and protein-reactive functional group (PRFG) diversity. FFSMPs contain: (1) a protein-binding motif, (2) an electrophilic or photoreactive PRFG for target protein capture, and (3) a terminal alkyne for click chemistry-based proteomic applications. These compounds can be directly applied in phenotypic screening programs to identify ligand-protein pairs in cells unbiasedly. Herein, we highlight 17 examples from 34 structurally diverse FFSMPs featuring five electrophiles, three photoreactive groups, and 15 chemical scaffolds. Essential to the synthesis of the FFSMPs was a new minimalist bifunctional isocyanide in an "isocyanide-based multicomponent reaction-Boc deprotection-arming" synthetic sequence. To the best of our knowledge, this is the first report concerning the preparation of appendage- and scaffold-diverse FFSMPs for integrated phenotypic screening-target identification campaigns with the ability to examine either electrophilic or photoreactive PRFGs. In contrast, the status quo for such studies has been appendage-diverse FFSMPs comprised of a single chemical scaffold and a single PRFG, which limits efficient target protein capture and/or chemical space sampling significantly in the quest for discovering new drug targets and/or compounds with novel mechanisms of action. Phenotypic screening of the electrophilic members of our library identified several FFSMPs with potent antiproliferative activity against MCF10CA1a breast cancer cells. One of these FFSMPs (Compound 4a) covalently targeted and potently inhibited protein disulfide isomerase A1 (PDIA1). This study supports the continued use of minimalist bifunctional isocyanides as valuable building blocks for preparing structurally diverse FFSMPs for integrated phenotypic screening-target identification campaigns.
Collapse
Affiliation(s)
- Paul A. Jackson
- Graduate
School of Pharmaceutical Sciences, Duquesne
University, 600 Forbes Avenue, Pittsburgh, Pennsylvania 15282, United States
| | - Eleni Kisty
- Department
of Chemistry, Boston College, Merkert Chemistry
Center, 2609 Beacon Street, Chestnut Hill, Massachusetts 02464, United States
| | - Vandan Pradhan
- Graduate
School of Pharmaceutical Sciences, Duquesne
University, 600 Forbes Avenue, Pittsburgh, Pennsylvania 15282, United States
| | - Christopher Swank
- Graduate
School of Pharmaceutical Sciences, Duquesne
University, 600 Forbes Avenue, Pittsburgh, Pennsylvania 15282, United States
| | - Luke Bohrer
- Graduate
School of Pharmaceutical Sciences, Duquesne
University, 600 Forbes Avenue, Pittsburgh, Pennsylvania 15282, United States
| | - Tammy L. Nolan
- Graduate
School of Pharmaceutical Sciences, Duquesne
University, 600 Forbes Avenue, Pittsburgh, Pennsylvania 15282, United States
| | - Eranthie Weerapana
- Department
of Chemistry, Boston College, Merkert Chemistry
Center, 2609 Beacon Street, Chestnut Hill, Massachusetts 02464, United States
| | - David J. Lapinsky
- Graduate
School of Pharmaceutical Sciences, Duquesne
University, 600 Forbes Avenue, Pittsburgh, Pennsylvania 15282, United States
| |
Collapse
|
28
|
Bálint D, Póti ÁL, Alexa A, Sok P, Albert K, Torda L, Földesi-Nagy D, Csókás D, Turczel G, Imre T, Szarka E, Fekete F, Bento I, Bojtár M, Palkó R, Szabó P, Monostory K, Pápai I, Soós T, Reményi A. Reversible covalent c-Jun N-terminal kinase inhibitors targeting a specific cysteine by precision-guided Michael-acceptor warheads. Nat Commun 2024; 15:8606. [PMID: 39366946 PMCID: PMC11452492 DOI: 10.1038/s41467-024-52573-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/13/2024] [Indexed: 10/06/2024] Open
Abstract
There has been a surge of interest in covalent inhibitors for protein kinases in recent years. Despite success in oncology, the off-target reactivity of these molecules is still hampering the use of covalent warhead-based strategies. Herein, we disclose the development of precision-guided warheads to mitigate the off-target challenge. These reversible warheads have a complex and cyclic structure with optional chirality center and tailored steric and electronic properties. To validate our proof-of-concept, we modified acrylamide-based covalent inhibitors of c-Jun N-terminal kinases (JNKs). We show that the cyclic warheads have high resilience against off-target thiols. Additionally, the binding affinity, residence time, and even JNK isoform specificity can be fine-tuned by adjusting the substitution pattern or using divergent and orthogonal synthetic elaboration of the warhead. Taken together, the cyclic warheads presented in this study will be a useful tool for medicinal chemists for the deliberate design of safer and functionally fine-tuned covalent inhibitors.
Collapse
Affiliation(s)
- Dániel Bálint
- Organocatalysis Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
- Hevesy György PhD School of Chemistry, Eötvös Loránd University, 1117, Budapest, Hungary
| | - Ádám Levente Póti
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
- Doctoral School of Biology, Eötvös Loránd University, 1117, Budapest, Hungary
| | - Anita Alexa
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Péter Sok
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Krisztián Albert
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Lili Torda
- Organocatalysis Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Dóra Földesi-Nagy
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Dániel Csókás
- Theoretical Chemistry Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Gábor Turczel
- NMR Research Laboratory, Centre for Structural Science, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Tímea Imre
- MS Metabolomic Research Laboratory, Centre for Structural Science, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Eszter Szarka
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Ferenc Fekete
- Metabolic Drug-interactions Research Group, Institute of Molecular Life Sciences, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Isabel Bento
- European Molecular Biology Laboratory, EMBL, Hamburg, Germany
| | - Márton Bojtár
- Chemical Biology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Roberta Palkó
- Organocatalysis Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Pál Szabó
- MS Metabolomic Research Laboratory, Centre for Structural Science, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Katalin Monostory
- Metabolic Drug-interactions Research Group, Institute of Molecular Life Sciences, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Imre Pápai
- Theoretical Chemistry Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Tibor Soós
- Organocatalysis Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary.
| | - Attila Reményi
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary.
| |
Collapse
|
29
|
Póti ÁL, Bálint D, Alexa A, Sok P, Ozsváth K, Albert K, Turczel G, Magyari S, Ember O, Papp K, Király SB, Imre T, Németh K, Kurtán T, Gógl G, Varga S, Soós T, Reményi A. Targeting a key protein-protein interaction surface on mitogen-activated protein kinases by a precision-guided warhead scaffold. Nat Commun 2024; 15:8607. [PMID: 39366929 PMCID: PMC11452651 DOI: 10.1038/s41467-024-52574-1] [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: 08/22/2023] [Accepted: 08/22/2024] [Indexed: 10/06/2024] Open
Abstract
For mitogen-activated protein kinases (MAPKs) a shallow surface-distinct from the substrate binding pocket-called the D(ocking)-groove governs partner protein binding. Screening of broad range of Michael acceptor compounds identified a double-activated, sterically crowded cyclohexenone moiety as a promising scaffold. We show that compounds bearing this structurally complex chiral warhead are able to target the conserved MAPK D-groove cysteine via reversible covalent modification and interfere with the protein-protein interactions of MAPKs. The electronic and steric properties of the Michael acceptor can be tailored via different substitution patterns. The inversion of the chiral center of the warhead can reroute chemical bond formation with the targeted cysteine towards the neighboring, but less nucleophilic histidine. Compounds bind to the shallow MAPK D-groove with low micromolar affinity in vitro and perturb MAPK signaling networks in the cell. This class of chiral, cyclic and enhanced 3D shaped Michael acceptor scaffolds offers an alternative to conventional ATP-competitive drugs modulating MAPK signaling pathways.
Collapse
Affiliation(s)
- Ádám Levente Póti
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
- Doctoral School of Biology, Eötvös Loránd University, Budapest, Hungary
| | - Dániel Bálint
- Organocatalysis Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
- Hevesy György PhD School of Chemistry, Eötvös Loránd University, Budapest, Hungary
| | - Anita Alexa
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | - Péter Sok
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | - Kristóf Ozsváth
- Organocatalysis Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | - Krisztián Albert
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | - Gábor Turczel
- NMR Research Laboratory, Centre for Structural Science, Research Centre for Natural Sciences, Budapest, Hungary
| | - Sarolt Magyari
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | - Orsolya Ember
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | - Kinga Papp
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | | | - Tímea Imre
- MS Metabolomic Research Laboratory, Centre for Structural Science, Research Centre for Natural Sciences, Budapest, Hungary
| | - Krisztina Németh
- MS Metabolomic Research Laboratory, Centre for Structural Science, Research Centre for Natural Sciences, Budapest, Hungary
| | - Tibor Kurtán
- Department of Organic Chemistry, University of Debrecen, Debrecen, Hungary
| | - Gergő Gógl
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | - Szilárd Varga
- Organocatalysis Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | - Tibor Soós
- Organocatalysis Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary.
| | - Attila Reményi
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary.
| |
Collapse
|
30
|
Liang L, Zhang Z, You Q, Guo X. Recent advances in the design of small molecular drugs with acrylamides covalent warheads. Bioorg Med Chem 2024; 112:117902. [PMID: 39236467 DOI: 10.1016/j.bmc.2024.117902] [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: 06/11/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024]
Abstract
In the development of covalent inhibitors, acrylamides warhead is one of the most popular classes of covalent warheads. In recent years, researchers have made different structural modifications to acrylamides warheads, resulting in the creation of fluorinated acrylamide warheads and cyano acrylamide warheads. These new warheads exhibit superior selectivity, intracellular accumulation, and pharmacokinetic properties. Additionally, although ketoamide warheads have been applied in the design of covalent inhibitors for viral proteins, it has not received sufficient attention. Combined with the studies in kinase inhibitors and antiviral drugs, this review presents the structural features and the progression of acrylamides warheads, offering a perspective on future research and development in this field.
Collapse
Affiliation(s)
- Luxia Liang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ze Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Xiaoke Guo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
31
|
Xu G, Torri D, Cuesta-Hoyos S, Panda D, Yates LRL, Zallot R, Bian K, Jia D, Iorgu AI, Levy C, Shepherd SA, Micklefield J. Cryptic enzymatic assembly of peptides armed with β-lactone warheads. Nat Chem Biol 2024; 20:1371-1379. [PMID: 38951647 PMCID: PMC11427300 DOI: 10.1038/s41589-024-01657-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 05/29/2024] [Indexed: 07/03/2024]
Abstract
Nature has evolved biosynthetic pathways to molecules possessing reactive warheads that inspired the development of many therapeutic agents, including penicillin antibiotics. Peptides armed with electrophilic warheads have proven to be particularly effective covalent inhibitors, providing essential antimicrobial, antiviral and anticancer agents. Here we provide a full characterization of the pathways that nature deploys to assemble peptides with β-lactone warheads, which are potent proteasome inhibitors with promising anticancer activity. Warhead assembly involves a three-step cryptic methylation sequence, which is likely required to reduce unfavorable electrostatic interactions during the sterically demanding β-lactonization. Amide-bond synthetase and adenosine triphosphate (ATP)-grasp enzymes couple amino acids to the β-lactone warhead, generating the bioactive peptide products. After reconstituting the entire pathway to β-lactone peptides in vitro, we go on to deliver a diverse range of analogs through enzymatic cascade reactions. Our approach is more efficient and cleaner than the synthetic methods currently used to produce clinically important warhead-containing peptides.
Collapse
Affiliation(s)
- Guangcai Xu
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Daniele Torri
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Sebastian Cuesta-Hoyos
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Deepanjan Panda
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Luke R L Yates
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Rémi Zallot
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Kehan Bian
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Dongxu Jia
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Andreea I Iorgu
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Colin Levy
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Sarah A Shepherd
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Jason Micklefield
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK.
| |
Collapse
|
32
|
Scholtes JF, Alhambra C, Carpino PA. Trends in covalent drug discovery: a 2020-23 patent landscape analysis focused on select covalent reacting groups (CRGs) found in FDA-approved drugs. Expert Opin Ther Pat 2024; 34:843-861. [PMID: 39219095 DOI: 10.1080/13543776.2024.2400175] [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: 04/16/2024] [Revised: 07/02/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Covalent drugs contain electrophilic groups that can react with nucleophilic amino acids located in the active sites of proteins, particularly enzymes. Recently, there has been considerable interest in using covalent drugs to target non-catalytic amino acids in proteins to modulate difficult targets (i.e. targeted covalent inhibitors). Covalent compounds contain a wide variety of covalent reacting groups (CRGs), but only a few of these CRGs are present in FDA-approved covalent drugs. AREAS COVERED This review summarizes a 2020-23 patent landscape analysis that examined trends in the field of covalent drug discovery around targets and organizations. The analysis focused on patent applications that were submitted to the World International Patent Organization and selected using a combination of keywords and structural searches based on CRGs present in FDA-approved drugs. EXPERT OPINION A total of 707 patent applications from >300 organizations were identified, disclosing compounds that acted at 71 targets. Patent application counts for five targets accounted for ~63% of the total counts (i.e. BTK, EGFR, FGFR, KRAS, and SARS-CoV-2 Mpro). The organization with the largest number of patent counts was an academic institution (Dana-Farber Cancer Institute). For one target, KRAS G12C, the discovery of new drugs was highly competitive (>100 organizations, 186 patent applications).
Collapse
|
33
|
Priyanka, Raymandal B, Mondal S. Native State Stabilization of Amyloidogenic Proteins by Kinetic Stabilizers: Inhibition of Protein Aggregation and Clinical Relevance. ChemMedChem 2024; 19:e202400244. [PMID: 38863235 DOI: 10.1002/cmdc.202400244] [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: 04/05/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/13/2024]
Abstract
Proteinopathies or amyloidoses are a group of life-threatening disorders that result from misfolding of proteins and aggregation into toxic insoluble amyloid aggregates. Amyloid aggregates have low clearance from the body due to the insoluble nature, leading to their deposition in various organs and consequent organ dysfunction. While amyloid deposition in the central nervous system leads to neurodegenerative diseases that mostly cause dementia and difficulty in movement, several other organs, including heart, liver and kidney are also affected by systemic amyloidoses. Regardless of the site of amyloid deposition, misfolding and structural alteration of the precursor proteins play the central role in amyloid formation. Kinetic stabilizers are an emerging class of drugs, which act like pharmacological chaperones to stabilize the native state structure of amyloidogenic proteins and to increase the activation energy barrier that is required for adopting a misfolded structure or conformation, ultimately leading to the inhibition of protein aggregation. In this review, we discuss the kinetic stabilizers that stabilize the native quaternary structure of transthyretin, immunoglobulin light chain and superoxide dismutase 1 that cause transthyretin amyloidoses, light chain amyloidosis and familial amyotrophic lateral sclerosis, respectively.
Collapse
Affiliation(s)
- Priyanka
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, Hauz Khas, New Delhi, Delhi, 110016, India
| | - Bitta Raymandal
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, Hauz Khas, New Delhi, Delhi, 110016, India
| | - Santanu Mondal
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, Hauz Khas, New Delhi, Delhi, 110016, India
| |
Collapse
|
34
|
Tsuruno A, Kamoshita S, Hosoya S, Sakurai K. Dichlorotriazine-based multivalent probe for selective affinity labeling of carbohydrate-binding proteins. Org Biomol Chem 2024; 22:7659-7663. [PMID: 39193651 DOI: 10.1039/d4ob01285b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
A new series of multivalent gold nanoparticle probes bearing different electrophilic groups were synthesized and their affinity labeling reactivities were evaluated. The dichlorotriazine group was identified as a useful protein-reactive label, allowing selective capture of a target protein at nanomolar probe concentrations.
Collapse
Affiliation(s)
- Ayaka Tsuruno
- Tokyo University of Agriculture and Technology, Department of Biotechnology and Life Science, Koganei-shi, Tokyo 184-8588, Japan.
| | - Shione Kamoshita
- Tokyo University of Agriculture and Technology, Department of Biotechnology and Life Science, Koganei-shi, Tokyo 184-8588, Japan.
| | - Shoichi Hosoya
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kaori Sakurai
- Tokyo University of Agriculture and Technology, Department of Biotechnology and Life Science, Koganei-shi, Tokyo 184-8588, Japan.
| |
Collapse
|
35
|
den Hollander LS, Beerkens BLH, Dekkers S, van Veldhoven JPD, Ortiz Zacarías NV, van der Horst C, Sieders EG, de Valk B, Wang J, IJzerman AP, van der Es D, Heitman LH. Labeling of CC Chemokine Receptor 2 with a Versatile Intracellular Allosteric Probe. ACS Chem Biol 2024; 19:2070-2080. [PMID: 39186040 PMCID: PMC11420878 DOI: 10.1021/acschembio.4c00439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Interest in affinity-based probes (AfBPs) as novel tools to interrogate G protein-coupled receptors (GPCRs) has gained traction in recent years. AfBPs represent an interesting and more versatile alternative to antibodies. In the present study, we report the development and validation of AfBPs that target the intracellular allosteric pocket of CCR2, a GPCR of interest for the development of therapies targeting autoimmune and inflammatory diseases and also cancer. Owing to the two-step labeling process of these CCR2 AfBPs through the incorporation of a click handle, we were successful in applying our most efficient probe in a variety of in vitro experiments and making use of multiple different detection techniques, such as SDS-PAGE and LC/MS-based proteomics. Collectively, this novel probe shows high selectivity, versatility, and applicability. Hence, this is a valuable alternative for CCR2-targeting antibodies and other traditional tool compounds and could aid in target validation and engagement in drug discovery.
Collapse
Affiliation(s)
- Lisa S den Hollander
- Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, Leiden 2333 CC, The Netherlands
| | - Bert L H Beerkens
- Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, Leiden 2333 CC, The Netherlands
| | - Sebastian Dekkers
- Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, Leiden 2333 CC, The Netherlands
| | - Jacobus P D van Veldhoven
- Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, Leiden 2333 CC, The Netherlands
| | - Natalia V Ortiz Zacarías
- Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, Leiden 2333 CC, The Netherlands
| | - Cas van der Horst
- Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, Leiden 2333 CC, The Netherlands
| | - Elisabeth G Sieders
- Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, Leiden 2333 CC, The Netherlands
| | - Bert de Valk
- Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, Leiden 2333 CC, The Netherlands
| | - Jianhui Wang
- Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, Leiden 2333 CC, The Netherlands
| | - Adriaan P IJzerman
- Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, Leiden 2333 CC, The Netherlands
| | - Daan van der Es
- Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, Leiden 2333 CC, The Netherlands
| | - Laura H Heitman
- Leiden Academic Centre for Drug Research, Division of Medicinal Chemistry, Leiden 2333 CC, The Netherlands
| |
Collapse
|
36
|
Zhao Z, Bourne PE. Advances in reversible covalent kinase inhibitors. Med Res Rev 2024. [PMID: 39287197 DOI: 10.1002/med.22084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 08/07/2024] [Accepted: 09/02/2024] [Indexed: 09/19/2024]
Abstract
Reversible covalent kinase inhibitors (RCKIs) are a class of novel kinase inhibitors attracting increasing attention because they simultaneously show the selectivity of covalent kinase inhibitors yet avoid permanent protein-modification-induced adverse effects. Over the last decade, RCKIs have been reported to target different kinases, including Atypical group of kinases. Currently, three RCKIs are undergoing clinical trials. Here, advances in RCKIs are reviewed to systematically summarize the characteristics of electrophilic groups, chemical scaffolds, nucleophilic residues, and binding modes. In so doing, we integrate key insights into privileged electrophiles, the distribution of nucleophiles, and hence effective design strategies for the development of RCKIs. Finally, we provide a further perspective on future design strategies for RCKIs, including those that target proteins other than kinases.
Collapse
Affiliation(s)
- Zheng Zhao
- School of Data Science, University of Virginia, Charlottesville, Virginia, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Philip E Bourne
- School of Data Science, University of Virginia, Charlottesville, Virginia, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
37
|
Heppner DE, Ogboo BC, Urul DA, May EW, Schaefer EM, Murkin AS, Gehringer M. Demystifying Functional Parameters for Irreversible Enzyme Inhibitors. J Med Chem 2024; 67:14693-14696. [PMID: 39115869 DOI: 10.1021/acs.jmedchem.4c01721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Affiliation(s)
- David E Heppner
- Department of Chemistry, The State University of New York at Buffalo. Buffalo, New York 14221, United States
| | - Blessing C Ogboo
- Department of Chemistry, The State University of New York at Buffalo. Buffalo, New York 14221, United States
| | - Daniel A Urul
- AssayQuant Technologies Inc., Marlboro, Massachusetts 01752, United States
| | - Earl W May
- AssayQuant Technologies Inc., Marlboro, Massachusetts 01752, United States
| | - Erik M Schaefer
- AssayQuant Technologies Inc., Marlboro, Massachusetts 01752, United States
| | - Andrew S Murkin
- Department of Chemistry, The State University of New York at Buffalo. Buffalo, New York 14221, United States
| | - Matthias Gehringer
- Division of Medicinal Chemistry, Institute of Biomedical Engineering, University Hospital Tübingen and Institute of Pharmaceutical Sciences, University of Tübingen. 72076 Tübingen, Germany
| |
Collapse
|
38
|
Cao D, Xi R, Li H, Zhang Z, Shi X, Li S, Jin Y, Liu W, Zhang G, Liu X, Dong S, Feng X, Wang F. Discovery of a Covalent Inhibitor of Pro-Caspase-1 Zymogen Blocking NLRP3 Inflammasome Activation and Pyroptosis. J Med Chem 2024; 67:15873-15891. [PMID: 39159426 DOI: 10.1021/acs.jmedchem.4c01558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Caspase-1 plays a central role in innate immunity, as its activation by inflammasomes induces the production of proinflammatory cytokines and pyroptosis. However, specific inhibition of the enzymatic activity of this protease is not effective in suppressing inflammation, owing to its enzyme-independent function. Herein, we identified a cyclohexenyl isothiocyanate compound (CIB-1476) that potently inhibited caspase-1 activity and suppressed the assembly and activation of the NLRP3 inflammasome and gasdermin-D-mediated pyroptosis. Mechanistically, CIB-1476 directly targeted pro-caspase-1 as an irreversible covalent inhibitor by binding to Cys285 and Cys397, resulting in more durable anti-inflammatory effects in the suppression of enzyme-dependent IL-1β production and enzyme-independent nuclear factor κB activation. Chemoproteomic profiling demonstrated the engagement of CIB-1476 with caspase-1. CIB-1476 showed potent therapeutic effects by suppressing inflammasome activation in mice, which was abolished in Casp1-/- mice. These results warrant further development of CIB-1476 along with its analogues as a novel strategy for caspase-1 inhibitors.
Collapse
Affiliation(s)
- Dongyi Cao
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- Department of Pharmacy, Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming 650500, China
| | - Ruiying Xi
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongye Li
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Zhonghui Zhang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 511400, China
| | - Xiaoke Shi
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shanshan Li
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yujie Jin
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wanli Liu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, Beijing Advanced Innovation Center for Structural Biology, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing 100084, China
| | - Guolin Zhang
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Xiaohua Liu
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Shunxi Dong
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Xiaoming Feng
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Fei Wang
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| |
Collapse
|
39
|
Schnaider L, Tan S, Singh PR, Capuano F, Scott AJ, Hambley R, Lu L, Yang H, Wallace EJ, Jo H, DeGrado WF. SuFEx Chemistry Enables Covalent Assembly of a 280-kDa 18-Subunit Pore-Forming Complex. J Am Chem Soc 2024; 146:25047-25057. [PMID: 39190920 DOI: 10.1021/jacs.4c07920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Proximity-enhanced chemical cross-linking is an invaluable tool for probing protein-protein interactions and enhancing the potency of potential peptide and protein drugs. Here, we extend this approach to covalently stabilize large macromolecular assemblies. We used SuFEx chemistry to covalently stabilize an 18-subunit pore-forming complex, CsgG:CsgF, consisting of nine CsgG membrane protein subunits that noncovalently associate with nine CsgF peptides. Derivatives of the CsgG:CsgF pore have been used for DNA sequencing, which places high demands on the structural stability and homogeneity of the complex. To increase the robustness of the pore, we designed and synthesized derivatives of CsgF-bearing sulfonyl fluorides, which react with CsgG in very high yield to form a covalently stabilized CsgG:CsgF complex. The resulting pores formed highly homogeneous channels when added to artificial membranes. The high yield and rapid reaction rate of the SuFEx reaction prompted molecular dynamics simulations, which revealed that the SO2F groups in the initially formed complex are poised for nucleophilic reaction with a targeted Tyr. These results demonstrate the utility of SuFEx chemistry to structurally stabilize very large (here, 280 kDa) assemblies.
Collapse
Affiliation(s)
- Lee Schnaider
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94143, United States
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94143, United States
| | - Sophia Tan
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94143, United States
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94143, United States
| | | | | | | | | | - Lei Lu
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94143, United States
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94143, United States
| | - Hyunjun Yang
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94143, United States
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94143, United States
| | | | - Hyunil Jo
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94143, United States
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94143, United States
| | - William F DeGrado
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94143, United States
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94143, United States
| |
Collapse
|
40
|
Huang W, Fayad E, Abu Ali OA, Qin HL. A portal to highly valuable indole-functionalized vinyl sulfonyl fluorides and allylic sulfonyl fluorides. Org Biomol Chem 2024; 22:7117-7120. [PMID: 39150283 DOI: 10.1039/d4ob01213e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
A practical and efficient method for the C-3 site selective alkenylation of indoles was developed for constructing novel indole-functionalized vinyl sulfonyl fluorides and indolyl allylic sulfonyl fluorides. The reaction is accomplished with exclusive regio- and stereoselectivity without using transition metal catalysts, providing novel products of great potential value in medicinal chemistry, chemical biology, and drug discovery.
Collapse
Affiliation(s)
- Wenzhuo Huang
- State Key Laboratory of Silicate Materials for Architectures; and School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China.
| | - Eman Fayad
- Department of Biotechnology, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Ola A Abu Ali
- Department of Chemistry, College of Science, Taif University, Taif 21944, Saudi Arabia.
| | - Hua-Li Qin
- State Key Laboratory of Silicate Materials for Architectures; and School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China.
| |
Collapse
|
41
|
Cerutti JP, Diniz LA, Santos VC, Vilchez Larrea SC, Alonso GD, Ferreira RS, Dehaen W, Quevedo MA. Structure-Aided Computational Design of Triazole-Based Targeted Covalent Inhibitors of Cruzipain. Molecules 2024; 29:4224. [PMID: 39275072 PMCID: PMC11396839 DOI: 10.3390/molecules29174224] [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: 07/03/2024] [Revised: 08/29/2024] [Accepted: 09/03/2024] [Indexed: 09/16/2024] Open
Abstract
Cruzipain (CZP), the major cysteine protease present in T. cruzi, the ethiological agent of Chagas disease, has attracted particular attention as a therapeutic target for the development of targeted covalent inhibitors (TCI). The vast chemical space associated with the enormous molecular diversity feasible to explore by means of modern synthetic approaches allows the design of CZP inhibitors capable of exhibiting not only an efficient enzyme inhibition but also an adequate translation to anti-T. cruzi activity. In this work, a computer-aided design strategy was developed to combinatorially construct and screen large libraries of 1,4-disubstituted 1,2,3-triazole analogues, further identifying a selected set of candidates for advancement towards synthetic and biological activity evaluation stages. In this way, a virtual molecular library comprising more than 75 thousand diverse and synthetically feasible analogues was studied by means of molecular docking and molecular dynamic simulations in the search of potential TCI of CZP, guiding the synthetic efforts towards a subset of 48 candidates. These were synthesized by applying a Cu(I)-catalyzed azide-alkyne cycloaddition (CuAAC) centered synthetic scheme, resulting in moderate to good yields and leading to the identification of 12 hits selectively inhibiting CZP activity with IC50 in the low micromolar range. Furthermore, four triazole derivatives showed good anti-T. cruzi inhibition when studied at 50 μM; and Ald-6 excelled for its high antitrypanocidal activity and low cytotoxicity, exhibiting complete in vitro biological activity translation from CZP to T. cruzi. Overall, not only Ald-6 merits further advancement to preclinical in vivo studies, but these findings also shed light on a valuable chemical space where molecular diversity might be explored in the search for efficient triazole-based antichagasic agents.
Collapse
Affiliation(s)
- Juan Pablo Cerutti
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC), Haya de la Torre y Medina Allende, Córdoba 5000, Argentina
- Sustainable Chemistry for Metals and Molecules, Department of Chemistry, KU Leuven, Celestijnenlaan 200F, 3001 Leuven, Belgium
| | - Lucas Abreu Diniz
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte 31270-901, Brazil
| | - Viviane Corrêa Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte 31270-901, Brazil
| | - Salomé Catalina Vilchez Larrea
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI-CONICET), Vuelta de Obligado 2490, Ciudad de Buenos Aires 1428, Argentina
| | - Guillermo Daniel Alonso
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI-CONICET), Vuelta de Obligado 2490, Ciudad de Buenos Aires 1428, Argentina
| | - Rafaela Salgado Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte 31270-901, Brazil
| | - Wim Dehaen
- Sustainable Chemistry for Metals and Molecules, Department of Chemistry, KU Leuven, Celestijnenlaan 200F, 3001 Leuven, Belgium
| | - Mario Alfredo Quevedo
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC), Haya de la Torre y Medina Allende, Córdoba 5000, Argentina
| |
Collapse
|
42
|
Kong L, Park SJ, Im W. CHARMM-GUI PDB Reader and Manipulator: Covalent Ligand Modeling and Simulation. J Mol Biol 2024; 436:168554. [PMID: 39237201 PMCID: PMC11377865 DOI: 10.1016/j.jmb.2024.168554] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 09/07/2024]
Abstract
Molecular modeling and simulation serve an important role in exploring biological functions of proteins at the molecular level, which is complementary to experiments. CHARMM-GUI (https://www.charmm-gui.org) is a web-based graphical user interface that generates complex molecular simulation systems and input files, and we have been continuously developing and expanding its functionalities to facilitate various complex molecular modeling and make molecular dynamics simulations more accessible to the scientific community. Currently, covalent drug discovery emerges as a popular and important field. Covalent drug forms a chemical bond with specific residues on the target protein, and it has advantages in potency for its prolonged inhibition effects. Even though there are higher demands in modeling PDB protein structures with various covalent ligand types, proper modeling of covalent ligands remains challenging. This work presents a new functionality in CHARMM-GUI PDB Reader & Manipulator that can handle a diversity of ligand-amino acid linkage types, which is validated by a careful benchmark study using over 1,000 covalent ligand structures in RCSB PDB. We hope that this new functionality can boost the modeling and simulation study of covalent ligands.
Collapse
Affiliation(s)
- Lingyang Kong
- Departments of Biological Sciences, Bioengineering, and Computer Science and Engineering, Lehigh University, Bethlehem, PA 18015, USA
| | - Sang-Jun Park
- Departments of Biological Sciences, Bioengineering, and Computer Science and Engineering, Lehigh University, Bethlehem, PA 18015, USA
| | - Wonpil Im
- Departments of Biological Sciences, Bioengineering, and Computer Science and Engineering, Lehigh University, Bethlehem, PA 18015, USA.
| |
Collapse
|
43
|
Chen X, Ding X, Fang J, Mao C, Gong X, Zhang Y, Zhang N, Yan F, Lou Y, Chen Z, Ding W, Ma Z. Natural Derivatives of Selective HDAC8 Inhibitors with Potent in Vivo Antitumor Efficacy against Breast Cancer. J Med Chem 2024; 67:14609-14632. [PMID: 39110628 DOI: 10.1021/acs.jmedchem.4c01438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
HDAC8 is a therapeutic target with great promise for breast cancer. Here, we reported a novel compound corallorazine D from Nocardiopsis sp. XZB108, selectively inhibited HDAC8 (IC50 = 0.90 ± 0.014 μM), suggesting that it may be a promising nonhydroxamate HDAC8 inhibitor. Upon additional modifications of corallorazine D, a candidate compound 5k, demonstrated remarkable inhibitory potency against HDAC8 (IC50 = 0.12 ± 0.01 nM), 89-fold superior to PCI-34051. The selectivity of 5k was at least 439-fold, superior to corallorazine D, confirming the efficacy of our modifications. In an orthotopic mouse model of breast cancer, 5k displayed nearly 4-fold superior antitumor activity than SAHA. Furthermore, 5k triggered antitumor immunity by activating T cells. Treatment with 5k significantly increased the proportion of M1 macrophages and decreased the proportion of M2 macrophages (M1/M2 ratio = 2.67 ± 0.25). 5k represents a promising compound for further investigation as a potential treatment for breast cancer.
Collapse
Affiliation(s)
- Xiaoming Chen
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Xia Ding
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Jiebin Fang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Churu Mao
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Xingzhi Gong
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Yuxiao Zhang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Ningjing Zhang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Feihang Yan
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Yijie Lou
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, the First Affiliated Hospital of Zhejiang Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhe Chen
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, the First Affiliated Hospital of Zhejiang Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Wanjing Ding
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
- Hainan Institute of Zhejiang University, Sanya 572025, China
| | - Zhongjun Ma
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
- Hainan Institute of Zhejiang University, Sanya 572025, China
| |
Collapse
|
44
|
Péczka N, Ranđelović I, Orgován Z, Csorba N, Egyed A, Petri L, Ábrányi-Balogh P, Gadanecz M, Perczel A, Tóvári J, Schlosser G, Takács T, Mihalovits LM, Ferenczy G, Buday L, Keserű GM. Contribution of Noncovalent Recognition and Reactivity to the Optimization of Covalent Inhibitors: A Case Study on KRas G12C. ACS Chem Biol 2024; 19:1743-1756. [PMID: 38991015 PMCID: PMC11334105 DOI: 10.1021/acschembio.4c00217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/23/2024] [Accepted: 07/01/2024] [Indexed: 07/13/2024]
Abstract
Covalent drugs might bear electrophiles to chemically modify their targets and have the potential to target previously undruggable proteins with high potency. Covalent binding of drug-size molecules includes a noncovalent recognition provided by secondary interactions and a chemical reaction leading to covalent complex formation. Optimization of their covalent mechanism of action should involve both types of interactions. Noncovalent and covalent binding steps can be characterized by an equilibrium dissociation constant (KI) and a reaction rate constant (kinact), respectively, and they are affected by both the warhead and the scaffold of the ligand. The relative contribution of these two steps was investigated on a prototypic drug target KRASG12C, an oncogenic mutant of KRAS. We used a synthetically more accessible nonchiral core derived from ARS-1620 that was equipped with four different warheads and a previously described KRAS-specific basic side chain. Combining these structural changes, we have synthesized novel covalent KRASG12C inhibitors and tested their binding and biological effect on KRASG12C by various biophysical and biochemical assays. These data allowed us to dissect the effect of scaffold and warhead on the noncovalent and covalent binding event. Our results revealed that the atropisomeric core of ARS-1620 is not indispensable for KRASG12C inhibition, the basic side chain has little effect on either binding step, and warheads affect the covalent reactivity but not the noncovalent binding. This type of analysis helps identify structural determinants of efficient covalent inhibition and may find use in the design of covalent agents.
Collapse
Affiliation(s)
- Nikolett Péczka
- Medicinal
Chemistry Research Group and National Drug Discovery and Development
Laboratory, HUN-REN Research Centre for
Natural Sciences, Budapest 1117, Hungary
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics, Budapest 1111, Hungary
| | - Ivan Ranđelović
- Department
of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest 1122, Hungary
| | - Zoltán Orgován
- Medicinal
Chemistry Research Group and National Drug Discovery and Development
Laboratory, HUN-REN Research Centre for
Natural Sciences, Budapest 1117, Hungary
| | - Noémi Csorba
- Medicinal
Chemistry Research Group and National Drug Discovery and Development
Laboratory, HUN-REN Research Centre for
Natural Sciences, Budapest 1117, Hungary
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics, Budapest 1111, Hungary
| | - Attila Egyed
- Medicinal
Chemistry Research Group and National Drug Discovery and Development
Laboratory, HUN-REN Research Centre for
Natural Sciences, Budapest 1117, Hungary
| | - László Petri
- Medicinal
Chemistry Research Group and National Drug Discovery and Development
Laboratory, HUN-REN Research Centre for
Natural Sciences, Budapest 1117, Hungary
| | - Péter Ábrányi-Balogh
- Medicinal
Chemistry Research Group and National Drug Discovery and Development
Laboratory, HUN-REN Research Centre for
Natural Sciences, Budapest 1117, Hungary
| | - Márton Gadanecz
- Protein
Modeling Research Group, Laboratory of Structural Chemistry and Biology, ELTE Institute of Chemistry, Budapest 1117, Hungary
- Hevesy
György PhD School of Chemistry, Eötvös
Loránd University, Pázmány Péter sétány. 1/A, Budapest 1117, Hungary
| | - András Perczel
- Protein
Modeling Research Group, Laboratory of Structural Chemistry and Biology, ELTE Institute of Chemistry, Budapest 1117, Hungary
| | - József Tóvári
- Department
of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest 1122, Hungary
| | - Gitta Schlosser
- MTA-ELTE
“Lendület”, Ion Mobility
Mass Spectrometry Research Group, Budapest 1117, Hungary
| | - Tamás Takács
- HUN-REN
Research Centre for Natural Sciences, Signal
Transduction and Functional Genomics Research Group, Budapest 1117, Hungary
- Doctoral
School of Biology, Institute of Biology, ELTE Eötvös Loránd University, Budapest 1117, Hungary
| | - Levente M. Mihalovits
- Medicinal
Chemistry Research Group and National Drug Discovery and Development
Laboratory, HUN-REN Research Centre for
Natural Sciences, Budapest 1117, Hungary
| | - György
G. Ferenczy
- Medicinal
Chemistry Research Group and National Drug Discovery and Development
Laboratory, HUN-REN Research Centre for
Natural Sciences, Budapest 1117, Hungary
| | - László Buday
- HUN-REN
Research Centre for Natural Sciences, Signal
Transduction and Functional Genomics Research Group, Budapest 1117, Hungary
| | - György M. Keserű
- Medicinal
Chemistry Research Group and National Drug Discovery and Development
Laboratory, HUN-REN Research Centre for
Natural Sciences, Budapest 1117, Hungary
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics, Budapest 1111, Hungary
| |
Collapse
|
45
|
Xu B, Zhang Z, Tantillo DJ, Dai M. Concise Total Syntheses of ( -)-Crinipellins A and B Enabled by a Controlled Cargill Rearrangement. J Am Chem Soc 2024; 146:21250-21256. [PMID: 39052841 PMCID: PMC11311239 DOI: 10.1021/jacs.4c07900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Herein, we report concise total syntheses of diterpene natural products (-)-crinipellins A and B with a tetraquinane skeleton, three adjacent all-carbon quaternary centers, and multiple oxygenated and labile functional groups. Our synthesis features a convergent Kozikowski β-alkylation to unite two readily available building blocks with all the required carbon atoms, an intramolecular photochemical [2 + 2] cycloaddition to install three challenging and adjacent all-carbon quaternary centers and a 5-6-4-5 tetracyclic skeleton, and a controlled Cargill rearrangement to rearrange the 5-6-4-5 tetracyclic skeleton to the desired tetraquinane skeleton. These strategically enabling transformations allowed us to complete total syntheses of (-)-crinipellins A and B in 12 and 13 steps, respectively. The results of quantum chemical computations revealed that the Bronsted acid-catalyzed Cargill rearrangements likely involve stepwise paths to products and the AlR3-catalyzed Cargill rearrangements likely involve a concerted path with asynchronous alkyl shifting events to form the desired product.
Collapse
Affiliation(s)
- Bo Xu
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Ziyao Zhang
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Dean J. Tantillo
- Department
of Chemistry, University of California—Davis, Davis, California 95616, United States
| | - Mingji Dai
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
- Department
of Pharmacology and Chemical Biology, Emory
University, Atlanta, Georgia 30322, United States
| |
Collapse
|
46
|
Rezaei E, Shahedi M, Habibi Z. Biocatalytic Synthesis of Nitrile-Bearing All-Carbon Quaternary Stereocenters. J Org Chem 2024; 89:10562-10571. [PMID: 39051740 DOI: 10.1021/acs.joc.4c00793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
The synthesis of all-carbon quaternary stereocenters containing nitriles is a very important and challenging subject in organic chemistry. We used a biocatalytic approach under mild conditions to obtain new derivatives of these scaffolds by oxidation of catechols by Myceliophthora thermophila laccase (Novozym 51003) to afford o-quinones and 1,4-addition of a series of carbon nucleophiles containing tertiary alkyle nitriles to these intermediates. Using this approach, α-cyano carbonyls bearing a quaternary stereocenter were also prepared. Finally, the yields for the prepared compounds were 72-94%.
Collapse
Affiliation(s)
- Elaheh Rezaei
- Department of Organic Chemistry, Shahid Beheshti University, 1983969411 Tehran, Iran
| | - Mansour Shahedi
- Department of Organic Chemistry, Shahid Beheshti University, 1983969411 Tehran, Iran
| | - Zohreh Habibi
- Department of Organic Chemistry, Shahid Beheshti University, 1983969411 Tehran, Iran
| |
Collapse
|
47
|
Lyu Y, Yang F, Sundaresh B, Rosconi F, van Opijnen T, Gao J. Covalent Inhibition of a Host-Pathogen Protein-Protein Interaction Reduces the Infectivity of Streptococcus pneumoniae. JACS AU 2024; 4:2484-2491. [PMID: 39055144 PMCID: PMC11267552 DOI: 10.1021/jacsau.4c00195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 07/27/2024]
Abstract
The ever-expanding antibiotic resistance urgently calls for novel antibacterial therapeutics, especially those with a new mode of action. We report herein our exploration of protein-protein interaction (PPI) inhibition as a new mechanism to thwart bacterial pathogenesis. Specifically, we describe potent and specific inhibitors of the pneumococcal surface protein PspC, an important virulence factor that facilitates the infection of Streptococcus pneumoniae. Specifically, PspC has been documented to recruit human complement factor H (hFH) to suppress host complement activation and/or promote the bacterial attachment to host tissues. The CCP9 domain of hFH was recombinantly expressed to inhibit the PspC-hFH interaction as demonstrated on live pneumococcal cells. The inhibitor allowed for the first pharmacological intervention of the PspC-hFH interaction. This PPI inhibition reduced pneumococci's attachment to epithelial cells and also resensitized the D39 strain of S. pneumoniae for opsonization. Importantly, we have further devised covalent versions of CCP9, which afforded long-lasting PspC inhibition with low nanomolar potency. Overall, our results showcase the promise of PPI inhibition for combating bacterial infections as well as the power of covalent inhibitors.
Collapse
Affiliation(s)
- Yuhan Lyu
- Department
of Chemistry, Merkert Chemistry Center, Boston College, Chestnut
Hill, Massachusetts 02467, United States
| | - Fan Yang
- Department
of Chemistry, Merkert Chemistry Center, Boston College, Chestnut
Hill, Massachusetts 02467, United States
| | - Bharathi Sundaresh
- Department
of Biology, Boston College, Chestnut Hill, Massachusetts 02467, United States
| | - Federico Rosconi
- Department
of Biology, Boston College, Chestnut Hill, Massachusetts 02467, United States
| | - Tim van Opijnen
- Broad
Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 02142, United States
| | - Jianmin Gao
- Department
of Chemistry, Merkert Chemistry Center, Boston College, Chestnut
Hill, Massachusetts 02467, United States
| |
Collapse
|
48
|
Liu Y, Li G, Ma W, Bao G, Li Y, He Z, Xu Z, Wang R, Sun W. Late-stage peptide modification and macrocyclization enabled by tertiary amine catalyzed tryptophan allylation. Chem Sci 2024; 15:11099-11107. [PMID: 39027288 PMCID: PMC11253200 DOI: 10.1039/d4sc01244e] [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: 02/21/2024] [Accepted: 06/11/2024] [Indexed: 07/20/2024] Open
Abstract
Late-stage modification of peptides could potentially endow peptides with significant bioactivity and physicochemical properties, and thereby provide novel opportunities for peptide pharmaceutical studies. Since tryptophan (Trp) bears a unique indole ring residue and plays various critical functional roles in peptides, the modification methods for tryptophan were preliminarily developed with considerable progress via transition-metal mediated C-H activation. Herein, we report an unprecedented tertiary amine catalyzed peptide allylation via the SN2'-SN2' pathway between the N1 position of the indole ring of Trp and Morita-Baylis-Hillman (MBH) carbonates. Using this method that proceeds under mild conditions, we demonstrated an extremely broad scope of Trp-containing peptides and MBH carbonates to prepare a series of peptide conjugates and cyclic peptides. The reaction is amenable to either solid-phase (on resin) or solution-phase conditions. In addition, the modified peptides can be further conjugated with other biomolecules at Trp, providing a new handle for bioconjugation.
Collapse
Affiliation(s)
- Yuyang Liu
- Research Unit of Peptide Science (2019RU066), Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College Beijing 100050 China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University Lanzhou 730000 China
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University Shenzhen 518055 China
| | - Guofeng Li
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University Shenzhen 518055 China
| | - Wen Ma
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University Lanzhou 730000 China
| | - Guangjun Bao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University Lanzhou 730000 China
| | - Yiping Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University Lanzhou 730000 China
| | - Zeyuan He
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University Lanzhou 730000 China
| | - Zhaoqing Xu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University Lanzhou 730000 China
| | - Rui Wang
- Research Unit of Peptide Science (2019RU066), Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College Beijing 100050 China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University Lanzhou 730000 China
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University Shenzhen 518055 China
| | - Wangsheng Sun
- Research Unit of Peptide Science (2019RU066), Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College Beijing 100050 China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University Lanzhou 730000 China
| |
Collapse
|
49
|
Lou J, Zhou Q, Lyu X, Cen X, Liu C, Yan Z, Li Y, Tang H, Liu Q, Ding J, Lu Y, Huang H, Xie H, Zhao Y. Discovery of a Covalent Inhibitor That Overcame Resistance to Venetoclax in AML Cells Overexpressing BFL-1. J Med Chem 2024; 67:10795-10830. [PMID: 38913996 DOI: 10.1021/acs.jmedchem.4c00291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Clinical and biological studies have shown that overexpression of BFL-1 is one contributing factor to venetoclax resistance. The resistance might be overcome by a potent BFL-1 inhibitor, but such an inhibitor is rare. In this study, we show that 56, featuring an acrylamide moiety, inhibited the BFL-1/BID interaction with a Ki value of 105 nM. More interestingly, 56 formed an irreversible conjugation adduct at the C55 residue of BFL-1. 56 was a selective BFL-1 inhibitor, and its MCL-1 binding affinity was 10-fold weaker, while it did not bind BCL-2 and BCL-xL. Mechanistic studies showed that 56 overcame venetoclax resistance in isogenic AML cell lines MOLM-13-OE and MV4-11-OE, which both overexpressed BFL-1. More importantly, 56 and venetoclax combination promoted stronger apoptosis induction than either single agent. Collectively, our data show that 56 overcame resistance to venetoclax in AML cells overexpressing BFL-1. These attributes make 56 a promising candidate for future optimization.
Collapse
MESH Headings
- Humans
- Sulfonamides/pharmacology
- Sulfonamides/chemistry
- Sulfonamides/chemical synthesis
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/chemistry
- Drug Resistance, Neoplasm/drug effects
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/chemical synthesis
- Cell Line, Tumor
- Minor Histocompatibility Antigens/metabolism
- Apoptosis/drug effects
- Drug Discovery
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Jianfeng Lou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd. Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Qianqian Zhou
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, PR China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd. Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xilin Lyu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd. Shanghai 201203, China
| | - Xinyi Cen
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Chen Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd. Shanghai 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ziqin Yan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd. Shanghai 201203, China
| | - Yan Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd. Shanghai 201203, China
| | - Haotian Tang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd. Shanghai 201203, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Qiupei Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd. Shanghai 201203, China
| | - Jian Ding
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, PR China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd. Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Ye Lu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd. Shanghai 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - He Huang
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, PR China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hua Xie
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, PR China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd. Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Yujun Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd. Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Shandong Provincial Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan 250101, China
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| |
Collapse
|
50
|
Raouf YS. Covalent Inhibitors: To Infinity and Beyond. J Med Chem 2024; 67:10513-10516. [PMID: 38913822 DOI: 10.1021/acs.jmedchem.4c01308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Affiliation(s)
- Yasir S Raouf
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain 15551, UAE
| |
Collapse
|