1
|
Qu X, Wang Y, Xu Y, Xu L, Ye X, Cai H, Bu L, Zeng Z, Zhou H. Aromatic nitroolefin with inhibition efficacy in triple-negative breast cancer cells by dual targeting RXRα and tubulins. Eur J Med Chem 2025; 289:117486. [PMID: 40090298 DOI: 10.1016/j.ejmech.2025.117486] [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: 01/07/2025] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/18/2025]
Abstract
We previously identified that 1-(2-Nitrovinyl)naphthalene (Z-10) is a ligand of retinoid x receptor α (RXRα) with a potent anti-breast cancer efficacy and revealed that nitro group is an essential pharmacophore in Z-10. In this study, we defined that the double bond of the nitrovinyl group is also vital for Z-10 to bind and activate RXRα. Mechanistically, the double bond has a chemical ability to mediate Z-10's covalent binding of RXRα via the Michael addition reaction with Cys432. By retaining the nitrovinyl group, a series of Z-10 analogues with different aromatic groups and different aromatic ring-positions of nitrovinyl group and alkoxy groups were designed and synthesized. We found that some analogues including compound 30 show stronger ability than Z-10 in inhibiting TNFα survival signal in MDA-MB-231 breast cancer cells. Interestingly, these RXRα ligands also bind to tubulins likely through the similar covalent interaction and induce the degradation of tubulins and cell cycle arrest in MDA-MB-231 cells, of which 30 displays the strongest efficacy. Importantly, these analogues and TNFα exhibit synergistic effects in inducing breast cancer cell apoptosis, of which 30 shows greater efficacy than Z-10. Together, our study provides a theoretical basis for the RXRα and tubulin dual-targeting drug design for breast cancer treatment.
Collapse
Affiliation(s)
- Xiaofang Qu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yanxia Wang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yunqing Xu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China
| | - Lin Xu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xiaohong Ye
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China; High Throughput Drug Screening Platform, Xiamen University, Xiamen, Fujian, 361102, China
| | - Hongchen Cai
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China
| | - Liang Bu
- Department of Thoracic Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China.
| | - Zhiping Zeng
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China; High Throughput Drug Screening Platform, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Hu Zhou
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China; High Throughput Drug Screening Platform, Xiamen University, Xiamen, Fujian, 361102, China.
| |
Collapse
|
2
|
Ramírez-Cortés SA, Durán-Vargas A, Rauda-Ceja JA, Mendoza-Espinosa P, Cofas-Vargas LF, Cruz-Rangel A, Pérez-Carreón JI, García-Hernández E. Targeting human prostaglandin reductase 1 with Licochalcone A: Insights from molecular dynamics and covalent docking studies. Biophys Chem 2025; 320-321:107410. [PMID: 39965292 DOI: 10.1016/j.bpc.2025.107410] [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: 09/13/2024] [Revised: 01/28/2025] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
Prostaglandin reductase 1 (PTGR1) is an NADPH-dependent enzyme critical to eicosanoid metabolism. Its elevated expression in malignant tumors often correlates with poor prognosis due to its role in protecting cells against reactive oxygen species. This study explores the inhibitory potential of licochalcone A, a flavonoid derived from Xinjiang licorice root, on human PTGR1. Using molecular dynamics simulations, we mapped the enzyme's conformational landscape, revealing a low-energy, rigid-body-like movement of the catalytic domain relative to the nucleotide-binding domain that governs PTGR1's transition between open and closed states. Simulations of NADPH-depleted dimer and NADPH-bound monomer highlighted the critical role of intersubunit interactions and coenzyme binding in defining PTGR1's conformational landscape, offering a deeper understanding of its functional adaptability as a holo-homodimer. Covalent docking, informed by prior chemoproteomic cross-linking data, revealed a highly favorable binding pose for licochalcone A at the NADPH-binding site. This pose aligned with a transient noncovalent binding pose inferred from solvent site-guided molecular docking, emphasizing the stereochemical complementarity of the coenzyme-binding site to licochalcone A. Sequence analysis across PTGR1 orthologs in vertebrates and exploration of 3D structures of human NADPH-binding proteins further underscore the potential of the coenzyme-binding site as a scaffold for developing PTGR1-specific inhibitors, positioning licochalcone A as a promising lead compound.
Collapse
Affiliation(s)
- Sara Abigail Ramírez-Cortés
- Universidad Nacional Autónoma de México, Instituto de Química, Ciudad Universitaria, Ciudad de Mexico, 04510, Mexico
| | - Adrián Durán-Vargas
- Universidad Nacional Autónoma de México, Instituto de Química, Ciudad Universitaria, Ciudad de Mexico, 04510, Mexico
| | - Jesús Antonio Rauda-Ceja
- Universidad Nacional Autónoma de México, Instituto de Química, Ciudad Universitaria, Ciudad de Mexico, 04510, Mexico
| | - Paola Mendoza-Espinosa
- Universidad Nacional Autónoma de México, Instituto de Química, Ciudad Universitaria, Ciudad de Mexico, 04510, Mexico
| | - Luis Fernando Cofas-Vargas
- Biosystems and Soft Matter Division, Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawińskiego 5B, 02-106 Warsaw, Poland
| | - Armando Cruz-Rangel
- Biochemistry of Chronic Diseases Laboratory, National Institute of Genomic Medicine, INMEGEN, Mexico City, Mexico
| | | | - Enrique García-Hernández
- Universidad Nacional Autónoma de México, Instituto de Química, Ciudad Universitaria, Ciudad de Mexico, 04510, Mexico.
| |
Collapse
|
3
|
Warner TC, Marando VM, Santiago-Reyes OA, Hart EM, Smelyansky SR, Carter AW, Bernhardt TG, Bryson BD, Kim DE, Kiessling LL. Intercepting a Mycobacterial Biosynthetic Pathway with Covalent Labeling. J Am Chem Soc 2025; 147:11189-11198. [PMID: 40126103 DOI: 10.1021/jacs.4c17913] [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: 03/25/2025]
Abstract
The mycobacterial cell envelope plays both infectious and protective roles. Understanding its structure is crucial for unlocking the molecular basis underlying these functions. Studying glycans, the primary components of the cell envelope, is challenging due to their limited native functional handles for chemoselective modification. New labeling methods exploit biorthogonal chemistry, using small molecule mimics that intercept cellular metabolism or late-stage glycan biosynthesis. However, these strategies can have practical limitations, including probe delivery and effectiveness. An ideal small molecule probe should be easily deployed and exploit the critical enzyme-substrate relationships of natural substrates. To this end, we developed a "probegenic" strategy to label mycobacteria. Our approach eliminates the need for explicit substrate mimicry, as the relevant functionality is revealed by a target enzyme. Specifically, we synthesized an azide-substituted trans-β-lactone probe (AzLac), which adopts a substrate-like structure upon covalent enzyme labeling. This probe is incorporated by mycolyltransferases into a core mycobacterial cell envelope glycan, including in the pathogen Mycobacterium tuberculosis. Unlike other probes of the cell envelope, AzLac facilitates selective covalent labeling of the inner leaflet of the mycomembrane. Using Corynebacterium glutamicum mycolyltransferase deletion strains, we implicated Cmt2 as the primary mycolyltransferase target. We leveraged the ability to modify the cell envelope by demonstrating that AzLac could be used to attach a DNA barcode to mycobacteria, which would help track infection dynamics. Thus, we expect AzLac will be a valuable means of monitoring and tracking the mycobacterial cell envelope. Moreover, we anticipate masking and revealing recognition motifs in probes can be applied to diverse cellular targets.
Collapse
Affiliation(s)
- Theodore C Warner
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Victoria M Marando
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Omar A Santiago-Reyes
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Elizabeth M Hart
- Department of Microbiology, Harvard University Medical School, Boston, Massachusetts 02115, United States
- Howard Hughes Medical Institute, Harvard University Medical School, Boston, Massachusetts 02115, United States
| | - Stephanie R Smelyansky
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Alan W Carter
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Thomas G Bernhardt
- Department of Microbiology, Harvard University Medical School, Boston, Massachusetts 02115, United States
- Howard Hughes Medical Institute, Harvard University Medical School, Boston, Massachusetts 02115, United States
| | - Bryan D Bryson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Daria E Kim
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Laura L Kiessling
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
4
|
Beckmann L, Liessmann F, Icker M, Rieger D, Schlegel P, Urban N, Schaefer M, Meiler J, Schoeder CT, Tretbar M. Identification and optimization of a small molecule inhibitor of the ovarian tumor protease of the Crimean-Congo hemorrhagic fever virus. Bioorg Med Chem 2025; 120:118093. [PMID: 39923558 DOI: 10.1016/j.bmc.2025.118093] [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: 12/12/2024] [Revised: 01/28/2025] [Accepted: 01/28/2025] [Indexed: 02/11/2025]
Abstract
Crimean-Congo hemorrhagic fever (CCHF) is a viral tick-borne disease with fatality rates of up to 30 %. Currently, there are no vaccines or specific antivirals available. The genome of the CCHF virus (CCHFV) encodes an ovarian tumor (OTU) protease with a deubiquitinating activity that is responsible for the evasion of the innate immune response. Therefore, the inhibition of the OTU protease could provide a strategy for the treatment of CCHFV infections. In this study, we screened for small-molecule inhibitors of CCHFV OTU using a fluorescent ubiquitin rhodamine 110 assay. We identified and validated a 2-aminothiazole hit compound (IC50 = 42.3 μM) followed by structure-activity relationships (SAR) studies resulting in a new inhibitor of the CCHFV OTU protease. The most active derivative is a competitive CCHFV OTU inhibitor with an IC50 value of 10.7 μM. Selectivity studies revealed that the ubiquitin-specific peptidase 7 (USP7), ubiquitin C-terminal hydrolase 5 (UCHL5), OTU deubiquitinase 1 (OTUD1), and Cezanne are also inhibited by this newly developed inhibitor indicating binding to conserved regions of the ubiquitin-binding site within the deubiquitinase superfamilies. Molecular docking into the active site of CCHFV OTU proposes starting points for further structural modifications to improve activity and selectivity. These structure-activity relationships are the first to our knowledge to be reported for the CCHFV OTU protease and will help guide further drug discovery efforts.
Collapse
Affiliation(s)
- Lorenz Beckmann
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University 04103 Leipzig, Germany
| | - Fabian Liessmann
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University 04103 Leipzig, Germany; Center for Scalable Data Analytics and Artificial Intelligence, Leipzig University 04105 Leipzig, Germany
| | - Maik Icker
- Institute for Organic Chemistry, Faculty of Chemistry and Mineralogy, Leipzig University 04103 Leipzig, Germany
| | - Dominic Rieger
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University 04103 Leipzig, Germany
| | - Phillip Schlegel
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University 04103 Leipzig, Germany
| | - Nicole Urban
- Rudolf-Boehm Institute for Pharmacology and Toxicology, Faculty of Medicine, Leipzig University 04107 Leipzig, Germany
| | - Michael Schaefer
- Rudolf-Boehm Institute for Pharmacology and Toxicology, Faculty of Medicine, Leipzig University 04107 Leipzig, Germany
| | - Jens Meiler
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University 04103 Leipzig, Germany; Center for Scalable Data Analytics and Artificial Intelligence, Leipzig University 04105 Leipzig, Germany; Institute for Computer Science, Wilhelm Ostwald Institute for Physical and Theoretical Chemistry, Leipzig University, Leipzig, Germany; School of Embedded Composite Artificial Intelligence SECAI, Dresden/Leipzig, Germany; Department of Chemistry, Department of Pharmacology, Center for Structural Biology, Institute of Chemical Biology, Center for Applied Artificial Intelligence in Protein Dynamics, Vanderbilt University, Nashville, TN, United States of America
| | - Clara T Schoeder
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University 04103 Leipzig, Germany; Center for Scalable Data Analytics and Artificial Intelligence, Leipzig University 04105 Leipzig, Germany.
| | - Maik Tretbar
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University 04103 Leipzig, Germany.
| |
Collapse
|
5
|
Lee J, Son S, Lee M, Park SB. Development of potential immunomodulatory ligands targeting natural killer T cells inspired by gut symbiont-derived glycolipids. Commun Chem 2025; 8:98. [PMID: 40169880 DOI: 10.1038/s42004-025-01497-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/20/2025] [Indexed: 04/03/2025] Open
Abstract
α-Galactosylceramide (α-GalCer) is a prototypical antigen recognized by natural killer T (NKT) cells, a subset of T cells crucial for immune regulation. Despite its significance, the complex structure-activity relationship of α-GalCer and its analogs remains poorly understood, particularly in defining the structural determinants of NKT cell responses. In this study, we designed and synthesized potential immunomodulatory ligands targeting NKT cells, inspired by glycolipids derived from the gut symbiont Bacteroides fragilis. A series of α-GalCer analogs with terminal iso-branched sphinganine backbones was developed through rational modification of the acyl chain. Our results identified the C3' hydroxyl group as a structural element that impairs glycolipid presentation by CD1d, as evidenced by reduced IL-2 secretion and weak competition with a potent CD1d ligand. Notably, among C3'-deoxy α-GalCer analogs, those containing an α-chloroacetamide group exhibited robust NKT cell activation with Th2 selectivity. Computational docking and mass spectrometry analyses further confirmed the substantial interaction of α-chloroacetamide analogs to CD1d. These findings underscore the potential of leveraging microbiota-derived glycolipid structures to selectively modulate NKT cell functions for therapeutic purposes.
Collapse
Affiliation(s)
- Jesang Lee
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Sumin Son
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Minha Lee
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Seung Bum Park
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
6
|
Kunishige R, Noguchi Y, Okamoto N, Li L, Ono A, Murata M, Kano F. Protein covariation networks for elucidating ferroptosis inducer mechanisms and potential synergistic drug targets. Commun Biol 2025; 8:480. [PMID: 40164758 DOI: 10.1038/s42003-025-07886-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 03/04/2025] [Indexed: 04/02/2025] Open
Abstract
In drug development, systematically characterizing a compound's mechanism of action (MoA), including its direct targets and effector proteins, is crucial yet challenging. Network-based approaches, unlike those focused solely on direct targets, effectively detect a wide range of cellular responses elicited by compounds. This study applied protein covariation network analysis, leveraging quantitative, morphological, and localization features from immunostained microscopic images, to elucidate the MoA of AX-53802, a novel ferroptosis inducer. From the candidate targets extracted through network analysis, GPX4 was verified as the direct target by validation experiments. Additionally, aggregates involving GPX4, TfR1, and F-actin were observed alongside iron reduction, suggesting a ferroptosis defense mechanism. Furthermore, combination therapies targeting GPX4 and FAK/Src were found to enhance cancer cell death, and MDM2, ezrin, and cortactin were identified as potential ferroptosis inhibitor targets. These findings highlight the effectiveness of network-based approaches in uncovering a compound's MoA and developing combination therapies for cancer.
Collapse
Affiliation(s)
- Rina Kunishige
- Multimodal Cell Analysis Collaborative Research Cluster, Institute of Science Tokyo, Yokohama-shi, Kanagawa, Japan
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan
| | - Yoshiyuki Noguchi
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan
- International Research Center for Neurointelligence, Institutes for Advanced Study, The University of Tokyo, Tokyo, Japan
| | | | - Lei Li
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan
| | - Akito Ono
- Axcelead Drug Discovery Partners, Inc., Fujisawa, Kanagawa, Japan
| | - Masayuki Murata
- Multimodal Cell Analysis Collaborative Research Cluster, Institute of Science Tokyo, Yokohama-shi, Kanagawa, Japan
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan
| | - Fumi Kano
- Multimodal Cell Analysis Collaborative Research Cluster, Institute of Science Tokyo, Yokohama-shi, Kanagawa, Japan.
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan.
- Cell Biology Center, Institute of Integrated Research, Institute of Science Tokyo, Yokohama-shi, Kanagawa, Japan.
| |
Collapse
|
7
|
Petri L, Gabizon R, Ferenczy GG, Péczka N, Egyed A, Ábrányi-Balogh P, Takács T, Keserű GM. Size-Dependent Target Engagement of Covalent Probes. J Med Chem 2025; 68:6616-6632. [PMID: 40099438 PMCID: PMC11956015 DOI: 10.1021/acs.jmedchem.5c00017] [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: 01/03/2025] [Revised: 02/28/2025] [Accepted: 03/07/2025] [Indexed: 03/19/2025]
Abstract
Labeling proteins with covalent ligands is finding increasing use in proteomics applications, including identifying nucleophilic residues amenable for labeling and in the development of targeted covalent inhibitors (TCIs). Labeling efficiency is measured by the covalent occupancy of the target or by biochemical activity. Here, we investigate how these observed quantities relate to the intrinsic parameters of complex formation, namely, noncovalent affinity and covalent reactivity, and to experimental conditions, including incubation time and ligand concentration. It is shown that target engagement is beneficially driven by noncovalent recognition for lead-like compounds, which are appropriate starting points for targeted covalent inhibitors owing to their easily detectable occupancy and fixed binding mode, facilitating optimization. In contrast, labeling by fragment-sized compounds is inevitably reactivity-driven as their small size limits noncovalent affinity. They are well-suited for exploring ligandable nucleophilic residues, while small fragments are less appropriate starting points for TCI development.
Collapse
Affiliation(s)
- László Petri
- Medicinal
Chemistry Research Group and National Drug Discovery and Development
Laboratory, HUN-REN Research Centre for
Natural Sciences, 2 Magyar
tudósok krt, Budapest 1117, Hungary
| | - Ronen Gabizon
- Department
of Chemical and Structural Biology, Weizmann
Institute of Science, Helen and Milton A. Kimmelman bldg, Rehovot 76100, Israel
| | - György G. Ferenczy
- Medicinal
Chemistry Research Group and National Drug Discovery and Development
Laboratory, HUN-REN Research Centre for
Natural Sciences, 2 Magyar
tudósok krt, Budapest 1117, Hungary
| | - Nikolett Péczka
- Medicinal
Chemistry Research Group and National Drug Discovery and Development
Laboratory, HUN-REN Research Centre for
Natural Sciences, 2 Magyar
tudósok krt, Budapest 1117, Hungary
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics, 8 Budafoki út, Budapest 1111, Hungary
| | - Attila Egyed
- Medicinal
Chemistry Research Group and National Drug Discovery and Development
Laboratory, HUN-REN Research Centre for
Natural Sciences, 2 Magyar
tudósok krt, Budapest 1117, Hungary
| | - Péter Ábrányi-Balogh
- Medicinal
Chemistry Research Group and National Drug Discovery and Development
Laboratory, HUN-REN Research Centre for
Natural Sciences, 2 Magyar
tudósok krt, Budapest 1117, Hungary
| | - Tamás Takács
- HUN-REN
Research Centre for Natural Sciences, Signal
Transduction and Functional Genomics Research Group, 2 Magyar tudósok krt, Budapest 1117, Hungary
- Doctoral
School of Biology, Institute of Biology,
ELTE Eötvös Loránd University, Pázmány Péter sétány
1/A, Budapest 1117, Hungary
| | - György M. Keserű
- Medicinal
Chemistry Research Group and National Drug Discovery and Development
Laboratory, HUN-REN Research Centre for
Natural Sciences, 2 Magyar
tudósok krt, Budapest 1117, Hungary
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics, 8 Budafoki út, Budapest 1111, Hungary
| |
Collapse
|
8
|
Yang J, Chang Y, Zhou K, Huang W, Tien JCY, Zhang P, Liu W, Zhou L, Zhou Y, Ren X, Mannan R, Mahapatra S, Zhang Y, Hamadeh R, Ervine G, Wang Z, Wang GX, Chinnaiyan AM, Ding K. Discovery of YJZ5118: A Potent and Highly Selective Irreversible CDK12/13 Inhibitor with Synergistic Effects in Combination with Akt Inhibition. J Med Chem 2025; 68:6718-6734. [PMID: 40080446 DOI: 10.1021/acs.jmedchem.5c00127] [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: 03/15/2025]
Abstract
Cyclin-dependent kinases 12 and 13 (CDK12/13) have emerged as promising therapeutic targets for castration-resistant prostate cancer (CRPC) and other human cancers. Despite the development of several CDK12/13 inhibitors, challenges remain in achieving an optimal balance of potency, selectivity and pharmacokinetic properties. Here, we report the discovery of YJZ5118, a novel, potent and highly selective covalent inhibitor of CDK12/13 with reasonable pharmacokinetic profiles. YJZ5118 effectively inhibited CDK12 and CDK13 with IC50 values of 39.5 and 26.4 nM, respectively, while demonstrating high selectivity over other CDKs. Mass spectrometry analysis, cocrystal structure determination, and pulldown-proteomic experiments confirmed the compound's covalent binding mode with CDK12/13. Functionally, YJZ5118 efficiently suppressed the transcription of DNA damage response genes, induced DNA damage, and triggered apoptosis. Moreover, the compound significantly inhibited the proliferation of multiple tumor cell lines, particularly prostate cancer cells. Notably, YJZ5118 exhibited synergistic effects with Akt inhibitors both in vitro and in vivo.
Collapse
Affiliation(s)
- Jianzhang Yang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Road, Shanghai 200032, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, 855 Xingye Avenue East, Guangzhou 511400, China
| | - Yu Chang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Kaijie Zhou
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Road, Shanghai 200032, China
| | - Weixue Huang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Road, Shanghai 200032, China
| | - Jean Ching-Yi Tien
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Pujuan Zhang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Road, Shanghai 200032, China
| | - Wenyan Liu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Road, Shanghai 200032, China
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Licheng Zhou
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, 855 Xingye Avenue East, Guangzhou 511400, China
| | - Yang Zhou
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, 855 Xingye Avenue East, Guangzhou 511400, China
| | - Xiaomei Ren
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Road, Shanghai 200032, China
| | - Rahul Mannan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Somnath Mahapatra
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yuping Zhang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Rudana Hamadeh
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Grafton Ervine
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Zhen Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Road, Shanghai 200032, China
| | - George Xiaoju Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Urology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ke Ding
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Road, Shanghai 200032, China
| |
Collapse
|
9
|
Cregg J, Pota K, Tomlinson ACA, Yano J, Marquez A, Liu Y, Schulze CJ, Seamon KJ, Holderfield M, Wei X, Zhuang Y, Yang YC, Jiang J, Huang Y, Zhao R, Ling Y, Wang Z, Flagella M, Wang Z, Singh M, Knox JE, Nichols R, Wildes D, Smith JAM, Koltun ES, Gill AL. Discovery of Elironrasib (RMC-6291), a Potent and Orally Bioavailable, RAS(ON) G12C-Selective, Covalent Tricomplex Inhibitor for the Treatment of Patients with RAS G12C-Addicted Cancers. J Med Chem 2025; 68:6041-6063. [PMID: 39993169 DOI: 10.1021/acs.jmedchem.4c02313] [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: 02/26/2025]
Abstract
The discovery of elironrasib (RMC-6291) represents a significant breakthrough in targeting the previously deemed undruggable GTP-bound, active KRASG12C. To target the active state of RAS (RAS(ON)) directly, we have employed an innovative tri-complex inhibitor (TCI) modality involving formation of a complex with an inhibitor, the intracellular chaperone protein CypA, and the target protein KRASG12C in its GTP-bound form. The resulting tri-complex inhibits oncogenic signaling, inducing tumor regressions across various preclinical models of KRASG12C mutant human cancers. Here we report structure-guided medicinal chemistry efforts that led to the discovery of elironrasib, a potent, orally bioavailable, RAS(ON) G12C-selective, covalent, tri-complex inhibitor. The investigational agent elironrasib is currently undergoing phase 1 clinical trials (NCT05462717, NCT06128551, NCT06162221), with preliminary data indicating clinical activity in patients who had progressed on first-generation inactive state-selective KRASG12C inhibitors.
Collapse
Affiliation(s)
- James Cregg
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Kristof Pota
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | | | - Jason Yano
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Abby Marquez
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Yang Liu
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | | | - Kyle J Seamon
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | | | - Xing Wei
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Yongxian Zhuang
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Yu Chi Yang
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Jingjing Jiang
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Yue Huang
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Ruiping Zhao
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Yun Ling
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Zhican Wang
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Michael Flagella
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Zhengping Wang
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Mallika Singh
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - John E Knox
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Robert Nichols
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - David Wildes
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | | | - Elena S Koltun
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Adrian L Gill
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| |
Collapse
|
10
|
Chen P, Wang L, Wang X, Sun J, Miao F, Wang Z, Yang F, Xiang M, Gu M, Li S, Zhang J, Yuan P, Lu X, Zhang ZM, Gao L, Yao SQ. Cell-Active, Arginine-Targeting Irreversible Covalent Inhibitors for Non-Kinases and Kinases. Angew Chem Int Ed Engl 2025; 64:e202422372. [PMID: 39778034 DOI: 10.1002/anie.202422372] [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: 11/17/2024] [Revised: 12/31/2024] [Accepted: 01/08/2025] [Indexed: 01/11/2025]
Abstract
Targeted covalent inhibitors (TCIs) play an essential role in the fields of kinase research and drug discovery. TCI strategies to target more common amino acid side-chains have yet to be demonstrated. Targeting other amino acids would also expand the pharmaceutical industry's toolbox for targeting other tough-to-drug proteins. We report herein a glyoxal-based, arginine-reactive strategy to generate potent and selective small-molecule TCIs of Mcl-1 (an important anti-apoptotic protein) by selectively targeting the conserved arginine (R263) in the protein. We further validated the generality of this strategy by developing glyoxal-based, irreversible covalent inhibitors of AURKA (a cancer-related kinase) that showed exclusive reactivity with a solvent-exposed arginine (R220) of this enzyme. We showed the resulting compounds were potent, selective and cell-active, capable of covalently engaging endogenous AURKA in MV-4-11 cells with long residence time. Finally, we showed the potential application of glyoxal-based TCIs in targeting an acquired drug-resistance mutant of ALK kinase (G1202R).
Collapse
Affiliation(s)
- Peng Chen
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Lu Wang
- School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, China
| | - Xuan Wang
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Traditional Chinese Medicine, School of Pharmacy, Jinan University
| | - Jie Sun
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
- Department of Pharmacy, Linyi People's Hospital, Linyi, 276000, China
| | - Fengfei Miao
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
| | - Zuqin Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 510632, China
| | - Fang Yang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 510632, China
| | - Menghua Xiang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Mingxi Gu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Shengrong Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 510632, China
| | - Jianzhong Zhang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Peiyan Yuan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Xiaoyun Lu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 510632, China
| | - Zhi-Min Zhang
- School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, China
| | - Liqian Gao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
| |
Collapse
|
11
|
Emanuelson C, Naro Y, Shade O, Liu M, Khare SD, Deiters A. Rational Design of Stapled Covalent Peptide Modifiers of Oncoprotein E6 from Human Papillomavirus. ACS Chem Biol 2025; 20:746-757. [PMID: 40063062 PMCID: PMC11934087 DOI: 10.1021/acschembio.4c00878] [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: 12/28/2024] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025]
Abstract
Human Papillomavirus (HPV) is linked to multiple cancers, most significantly cervical cancer, for which HPV infection is associated with nearly all cases. Essential to the oncogenesis of HPV is the function of the viral protein E6 and its role in degrading the cell cycle regulator p53. Degradation of p53, and the resultant loss of cell cycle control, is mediated by E6 recruitment of the E3 ubiquitin ligase E6AP and subsequent ubiquitination of p53. Here, we report the design of a stapled peptide that mimics the LxxLL α-helical domain of E6AP to bind and covalently label a cysteine residue specific to HPV-16 E6. Several acrylamide- and haloacetamide-based warheads were evaluated for reactivity and specificity, and a panel of hydrocarbon-stapled peptides was evaluated for enhanced binding affinity and increased proteolytic stability. Structure-based modeling was used to rationalize the observed trends in the reactivity of the warheads and the impact of the hydrocarbon staple position on the binding affinity of the stapled peptides. The development of a proteolytically stable and reactive peptide represents a new class of peptide-based inhibitors of protein-protein interactions with a potential therapeutic value toward HPV-derived cancers.
Collapse
Affiliation(s)
- Cole Emanuelson
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Yuta Naro
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Olivia Shade
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Melinda Liu
- Department
of Chemistry and Chemical Biology, Rutgers
University, Piscataway, New Jersey 08854, United States
| | - Sagar D. Khare
- Department
of Chemistry and Chemical Biology, Rutgers
University, Piscataway, New Jersey 08854, United States
| | - Alexander Deiters
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
12
|
Shen R, Zhang B, Zhao L, Chang B, Zhang F, Chen Y, Fang J. A tunable stimuli-responsive module based on an α-hydroxymethyl-α,β-unsaturated carbonyl scaffold. J Mater Chem B 2025; 13:3980-3989. [PMID: 40029635 DOI: 10.1039/d4tb02818j] [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: 03/05/2025]
Abstract
The α-hydroxymethyl-α,β-unsaturated carbonyl (HMUC) scaffold represents a valuable framework for constructing nucleophile-responsive materials. However, nucleophiles are largely limited to thiols and amines. Given the ubiquity of thiols and amines in biological systems, this limitation hinders the creation of materials that can be selectively activated by exogenous stimuli. By tuning the electron density of the double bond and assessing its reactivity with various nucleophiles, we present here the discovery of the N-ethyl-2-(hydroxymethyl)acrylamide (NEHMAA) scaffold as a versatile building block for fabricating exogenous stimuli-responsive materials. The selenol species 4-cyanobenzylselenol (from its precursor bis(4-cyanobenzyl)diselenide, Se4) effectively activates NEHMAA-decorated "caged" molecules. Furthermore, the NEHMAA unit was employed to prepare prodrugs, and Se4-dependent cytotoxicity of these prodrugs was observed in cancer cells. The orthogonal reactivity between the NEHMAA unit and Se4 enriches the existing repertoire for constructing exogenous stimuli-responsive smart materials.
Collapse
Affiliation(s)
- Ruipeng Shen
- School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing, Jiangsu 210094, China.
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Baoxin Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Lanning Zhao
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Bingbing Chang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Fang Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Yating Chen
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Jianguo Fang
- School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing, Jiangsu 210094, China.
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| |
Collapse
|
13
|
Jin Y, Jana S, Abbasov ME, Lin H. Antibiotic target discovery by integrated phenotypic and activity-based profiling of electrophilic fragments. Cell Chem Biol 2025; 32:434-448.e9. [PMID: 40020665 DOI: 10.1016/j.chembiol.2025.02.001] [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: 04/27/2024] [Revised: 10/05/2024] [Accepted: 02/06/2025] [Indexed: 03/03/2025]
Abstract
The emergence of antibiotic resistance necessitates the discovery of novel bacterial targets and antimicrobial agents. Here, we present a bacterial target discovery framework that integrates phenotypic screening of cysteine-reactive fragments with competitive activity-based protein profiling to map and functionally characterize the targets of screening hits. Using this approach, we identify β-ketoacyl-acyl carrier protein synthase III (FabH) and MiaA tRNA prenyltransferase as primary targets of a hit fragment, 10-F05, that confer bacterial stress resistance and virulence in Shigella flexneri. Mechanistic investigations elucidate that covalent C112 modification in FabH, an enzyme involved in bacterial fatty acid synthesis, results in its inactivation and consequent growth inhibition. We further demonstrate that irreversible C273 modification at the MiaA RNA-protein interaction interface abrogates substrate tRNA binding, attenuating resistance and virulence through decreased translational accuracy. Our findings underscore the efficacy of integrating phenotypic and activity-based profiling of electrophilic fragments to accelerate the identification and pharmacologic validation of new therapeutic targets.
Collapse
Affiliation(s)
- Yizhen Jin
- Graduate Program of Biochemistry, Molecular and Cell Biology, Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA; Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA; Department of Medicine and Department of Chemistry, The University of Chicago, 900 E. 57(th) Street, Chicago, IL 60637, USA
| | - Sadhan Jana
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA; Department of Medicine and Department of Chemistry, The University of Chicago, 900 E. 57(th) Street, Chicago, IL 60637, USA
| | - Mikail E Abbasov
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA.
| | - Hening Lin
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA; Howard Hughes Medical Institute, Department of Medicine and Department of Chemistry, The University of Chicago, 900 E. 57(th) Street, Chicago, IL 60637, USA.
| |
Collapse
|
14
|
Paolino M, Tassone G, Governa P, Saletti M, Lami M, Carletti R, Sacchetta F, Pozzi C, Orlandini M, Manetti F, Olivucci M, Cappelli A. Morita-Baylis-Hillman Adduct Chemistry as a Tool for the Design of Lysine-Targeted Covalent Ligands. ACS Med Chem Lett 2025; 16:397-405. [PMID: 40104796 PMCID: PMC11912265 DOI: 10.1021/acsmedchemlett.4c00479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
The use of Targeted Covalent Inhibitors (TCIs) is an expanding strategy for the development of innovative drugs. It is driven by two fundamental steps: (1) recognition of the target site by the molecule and (2) establishment of the covalent interaction by its reactive group. The development of new TCIs depends on the development of new warheads. Here, we propose the use of Morita-Baylis-Hillman adducts (MBHAs) to covalently bind Lys strategically placed inside a lipophilic pocket. A human cellular retinoic acid binding protein II mutant (M2) was selected as a test bench for a library of 19 MBHAs. The noncovalent interaction step was investigated by molecular docking studies, while experimentally the entire library was incubated with M2 and crystallized to confirm covalent binding with the target lysine. The results, rationalized through covalent docking analysis, support our hypothesis of MBHAs as reactive scaffolds for the design of lysine-TCIs.
Collapse
Affiliation(s)
- Marco Paolino
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Giusy Tassone
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Paolo Governa
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Mario Saletti
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Matteo Lami
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Riccardo Carletti
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Filippo Sacchetta
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Cecilia Pozzi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Maurizio Orlandini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Fabrizio Manetti
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Massimo Olivucci
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
- Chemistry Department, Bowling Green State University, Overman Hall, Bowling Green, Ohio 43403, United States
| | - Andrea Cappelli
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| |
Collapse
|
15
|
Schultz CR, Aleiwi B, Zhou XE, Suino-Powell K, Melcher K, Almeida NMS, Wilson AK, Ellsworth EL, Bachmann AS. Design, Synthesis, and Biological Activity of Novel Ornithine Decarboxylase (ODC) Inhibitors. J Med Chem 2025; 68:5760-5773. [PMID: 40035393 PMCID: PMC11912471 DOI: 10.1021/acs.jmedchem.4c03120] [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: 12/18/2024] [Revised: 01/31/2025] [Accepted: 02/24/2025] [Indexed: 03/05/2025]
Abstract
We here describe the design, synthesis, and biological activity of novel ornithine decarboxylase (ODC) inhibitors that show significantly higher potency in vitro than α-difluoromethylornithine (DFMO), a U.S. Food and Drug Administration (FDA) approved drug. We report two X-ray structures of ODC complexed with new ODC inhibitors, computational docking, molecular dynamics, and binding free energy calculations to validate the experimental models. The X-ray structures reveal that covalent adducts with pyridoxal phosphate (PLP) are formed in the active site of the human ODC enzyme, as verified by their preparation and enzymatic testing. Finally, we verified that the cellular activity of endogenous ODC was inhibited, and polyamine levels were reduced. Given that ODC is a clinically validated target, combined with the fact that DFMO is currently the only ODC inhibitor in clinical use for several indications, the further development of more potent ODC inhibitors with superior activity and physical properties is warranted.
Collapse
Affiliation(s)
- Chad R Schultz
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, Michigan 49503, United States
- International Center for Polyamine Disorders, Grand Rapids, Michigan 49503, United States
| | - Bilal Aleiwi
- Department of Pharmacology and Toxicology, College of Human Medicine, East Lansing, Michigan 48824, United States
| | - X Edward Zhou
- Department of Structural Biology, Van Andel Institute, Grand Rapids, Michigan 49503, United States
| | - Kelly Suino-Powell
- Department of Structural Biology, Van Andel Institute, Grand Rapids, Michigan 49503, United States
| | - Karsten Melcher
- Department of Structural Biology, Van Andel Institute, Grand Rapids, Michigan 49503, United States
| | - Nuno M S Almeida
- Department of Chemistry, College of Natural Science, Michigan State University, East Lansing, Michigan 48824, United States
| | - Angela K Wilson
- Department of Chemistry, College of Natural Science, Michigan State University, East Lansing, Michigan 48824, United States
| | - Edmund L Ellsworth
- Department of Pharmacology and Toxicology, College of Human Medicine, East Lansing, Michigan 48824, United States
| | - André S Bachmann
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, Michigan 49503, United States
- International Center for Polyamine Disorders, Grand Rapids, Michigan 49503, United States
| |
Collapse
|
16
|
Niu GH, Hsiao WC, Lee PH, Zheng LG, Yang YS, Huang WC, Hsieh CC, Chiu TY, Wang JY, Chen CP, Huang CL, You MS, Kuo YP, Wang CM, Wen ZH, Yu GY, Chen CT, Chi YH, Tung CW, Hsu SC, Yeh TK, Sung PJ, Zhang MM, Tsou LK. Orally Bioavailable and Site-Selective Covalent STING Inhibitor Derived from a Macrocyclic Marine Diterpenoid. J Med Chem 2025; 68:5471-5487. [PMID: 40014799 PMCID: PMC11912488 DOI: 10.1021/acs.jmedchem.4c02665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/16/2025] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
Pharmacological inhibition of the cGAS-STING-controlled innate immune pathway is an emerging therapeutic strategy for a myriad of inflammatory diseases. Here, we report GHN105 as an orally bioavailable covalent STING inhibitor. Late-stage diversification of the briarane-type diterpenoid excavatolide B allowed the installation of solubility-enhancing functional groups while enhancing its activity as a covalent STING inhibitor against multiple human STING variants, including the S154 variant responsible for a genetic autoimmune disease. Selectively engaging the membrane-proximal Cys91 residue of STING, GHN105 dose-dependently inhibited cGAS-STING signaling and type I interferon responses in cells and in vivo. Moreover, orally administered GHN105 exhibited on-target engagement in vivo and markedly reversed key pathological features in a delayed treatment of the acute colitis mouse model. Our study provided proof of concept that the synthetic briarane analog GHN105 serves as a safe, site-selective, and orally active covalent STING inhibitor and devises a regimen that allows long-term systemic administration.
Collapse
Affiliation(s)
- Guang-Hao Niu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Wan-Chi Hsiao
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Po-Hsun Lee
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Li-Guo Zheng
- National Museum of Marine Biology and Aquarium, Pingtung 944401, Taiwan
| | - Yu-Shao Yang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Wei-Cheng Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Chih-Chien Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Tai-Yu Chiu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Jing-Ya Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Ching-Ping Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Chen-Lung Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - May-Su You
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Yi-Ping Kuo
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Chien-Ming Wang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung 804201, Taiwan
| | - Guann-Yi Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Ya-Hui Chi
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Chun-Wei Tung
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Shu-Ching Hsu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Ping-Jyun Sung
- National Museum of Marine Biology and Aquarium, Pingtung 944401, Taiwan
| | - Mingzi M Zhang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Lun Kelvin Tsou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| |
Collapse
|
17
|
He J, Illingworth H, Ullrich S, Ghosh P, Ton J, Jackson CJ, Nitsche C. Synthesis, screening and validation of cysteine-reactive fragments as chikungunya virus protease inhibitors. Bioorg Med Chem Lett 2025; 122:130176. [PMID: 40081602 DOI: 10.1016/j.bmcl.2025.130176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/21/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Alphaviruses like the Chikungunya virus cause severe outbreaks; however, no specific treatments are available. Their viral replication depends on the nsP2 cysteine protease, a promising but underexplored target for drug discovery. In this study, we report a covalent fragment screening against Chikungunya virus nsP2 protease, resulting in the identification of three inhibitors that can serve as starting points for future drug development. Careful validation proved indispensable in eliminating false-positive hits from a Förster resonance energy transfer (FRET)-based inhibition assay, wherein interference was caused by the inner filter effect between the fluorescent substrate and coloured compounds. Jump-dilution experiments accompanied by reactivity studies with cysteine and the recombinant protein indicate covalent inhibition via thia-Michael addition. We further demonstrate cross-inhibition of the related alphavirus nsP2 protease from Sindbis virus. The study provides early insights into nsP2 inhibition by electrophilic fragments featuring non-promiscuous N-arylacrylamides, thus advancing the search for antivirals targeting Chikungunya and other alphaviruses of concern.
Collapse
Affiliation(s)
- Junming He
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Henry Illingworth
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Sven Ullrich
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Pritha Ghosh
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Jennifer Ton
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Colin J Jackson
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia; Australian Research Council Centre of Excellence in Synthetic Biology, Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Christoph Nitsche
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia.
| |
Collapse
|
18
|
Zhuang J, Yang W, Cheng GJ. Mechanistic Insights into the Reversible Inhibition of d-Cycloserine in Alanine Racemase from Mycobacterium tuberculosis. J Chem Inf Model 2025; 65:2610-2622. [PMID: 39996595 DOI: 10.1021/acs.jcim.4c01763] [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: 02/26/2025]
Abstract
d-Cycloserine (DCS), an antibiotic used in the treatment of drug-resistant tuberculosis, was traditionally believed to irreversibly inhibit the pyridoxal-5'-phosphate (PLP)-dependent alanine racemase from Mycobacterium tuberculosis (MtAlr). However, recent research suggests that the inhibition is reversible, as MtAlr can be reactivated by destructing DCS. This study employs the hybrid quantum mechanics/molecular mechanics (QM/MM) method to investigate the mechanisms of MtAlr inhibition and DCS destruction. Computational results indicate that the inhibition reaction via an "isoxazole-forming" pathway is kinetically favorable, while the DCS destruction reaction via an "oxime-forming" pathway is thermodynamically favorable, explaining the irreversible inhibition of DCS. For the inhibition reaction, the isoxazole product was found to prefer the keto form, contrary to the previously proposed enol form. Moreover, K44 and D322' were identified as key residues. K44 transfers the proton from Cα and Cβ of DCS, while D322' stabilizes the carbanion intermediate and isoxazole product via electrostatic interaction with the protonated K44. Such electrostatic interaction was eliminated in the DCS-resistance variant, D322'N, making the inhibition reaction unfavorable. For DCS destruction, an "up-to-down" conformational change is required to place the isoxazolidinone ring in an appropriate position for hydrolysis. The deprotonated Y273' facilitates the hydrolysis reaction by enhancing the nucleophilicity of the water molecule. Throughout the whole reaction of MtAlr, PLP plays multiple roles, including stabilizing the carbanion intermediate and acting as a proton shuttle. Overall, this study provides deeper insight into the catalytic mechanism of MtAlr and offers valuable insights for drug development.
Collapse
Affiliation(s)
- Jingyuan Zhuang
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Wei Yang
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen 518112, China
| | - Gui-Juan Cheng
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| |
Collapse
|
19
|
Wang S, Faucher FF, Bertolini M, Kim H, Yu B, Cao L, Roeltgen K, Lovell S, Shanker V, Boyd SD, Wang L, Bartenschlager R, Bogyo M. Identification of Covalent Cyclic Peptide Inhibitors Targeting Protein-Protein Interactions Using Phage Display. J Am Chem Soc 2025; 147:7461-7475. [PMID: 39993812 DOI: 10.1021/jacs.4c15843] [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: 02/26/2025]
Abstract
Peptide macrocycles are promising therapeutics for a variety of disease indications due to their overall metabolic stability and potential to make highly selective binding interactions with targets. Recent advances in covalent macrocycle peptide discovery, driven by phage and mRNA display methods, have enabled the rapid identification of highly potent and selective molecules from large libraires of diverse macrocycles. However, there are currently limited examples of macrocycles that can be used to disrupt protein-protein interactions and even fewer examples that function by formation of a covalent bond to a target protein. In this work, we describe a directed counter-selection method that enables identification of covalent macrocyclic ligands targeting a protein-protein interaction using a phage display screening platform. This method utilizes binary and ternary screenings of a chemically modified phage display library, employing the stable and weakly reactive aryl fluorosulfate electrophile. We demonstrate the utility of this approach using the SARS-CoV-2 spike-ACE2 protein-protein interaction and identify multiple covalent macrocyclic inhibitors that disrupt this interaction. The resulting compounds displayed antiviral activity against live virus that was irreversible after washout due to the covalent binding mechanism. These results highlight the potential of this screening platform for developing covalent macrocyclic drugs that disrupt protein-protein interactions with long lasting effects.
Collapse
Affiliation(s)
- Sijie Wang
- Department of Pathology, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Franco F Faucher
- Department of Chemistry, School of Humanities and Sciences, Stanford University, Stanford, California 94305, United States
| | - Matilde Bertolini
- Department of Genetics, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Heeyoung Kim
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Diseases Research, Heidelberg University, Heidelberg 69210, Germany
| | - Bingchen Yu
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, California 94158, United States
| | - Li Cao
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, California 94158, United States
| | - Katharina Roeltgen
- Department of Pathology, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Scott Lovell
- Department of Pathology, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Varun Shanker
- Department of Biochemistry, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Scott D Boyd
- Department of Pathology, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Lei Wang
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, California 94158, United States
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Diseases Research, Heidelberg University, Heidelberg 69210, Germany
- Division Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
- German Center for Infection Research, Heidelberg Partner Site, Heidelberg 69120, Germany
| | - Matthew Bogyo
- Department of Pathology, School of Medicine, Stanford University, Stanford, California 94305, United States
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, United States
| |
Collapse
|
20
|
Rubicondo M, Ciardelli G, Mattu C, Tuszynski JA. Recent advancements in colchicine derivatives: Exploring synthesis, activities, and nanoformulations for enhanced therapeutic efficacy. Drug Discov Today 2025; 30:104312. [PMID: 39947582 DOI: 10.1016/j.drudis.2025.104312] [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: 11/07/2024] [Revised: 01/31/2025] [Accepted: 02/07/2025] [Indexed: 02/22/2025]
Abstract
The multifaceted anti-cancer properties of colchicine make it a promising candidate for tumor treatment. However, its application has been limited by poor solubility, low bioavailability, and systemic toxicity. Considerable efforts have been directed toward the development of colchicine derivatives and nanoformulations to overcome these challenges. In this review, we provide a comprehensive overview of recent advances in colchicine derivatives and nanoformulations for cancer treatment. Synthesis methods and in vitro antiproliferative assays for the reviewed derivatives and formulations are explored. Challenges, such as drug resistance and formulation optimization, are also addressed, along with future perspectives for leveraging the full potential of colchicine derivatives and their nanoformulations as innovative anti-cancer strategies, toward successful clinical applications.
Collapse
Affiliation(s)
- Marialucia Rubicondo
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; Polito BIOMed Lab, Politecnico di Torino, Turin, Italy
| | - Gianluca Ciardelli
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; Polito BIOMed Lab, Politecnico di Torino, Turin, Italy
| | - Clara Mattu
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; Polito BIOMed Lab, Politecnico di Torino, Turin, Italy
| | - Jack A Tuszynski
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada.
| |
Collapse
|
21
|
Wang S, Woods EC, Jo J, Zhu J, Hansel-Harris A, Holcomb M, Llanos M, Pedowitz NJ, Upadhyay T, Bennett J, Fellner M, Park KW, Zhang A, Valdez TA, Forli S, Chan AI, Cunningham CN, Bogyo M. An mRNA Display Approach for Covalent Targeting of a Staphylococcus aureus Virulence Factor. J Am Chem Soc 2025. [PMID: 40013487 DOI: 10.1021/jacs.4c15713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Staphylococcus aureus (S. aureus) is an opportunistic human pathogen that causes over one million deaths around the world each year. We recently identified a family of serine hydrolases termed fluorophosphonate binding hydrolases (Fphs) that play important roles in lipid metabolism and colonization of a host. Because many of these enzymes are only expressed in Staphylococcus bacteria, they are valuable targets for diagnostics and therapeutics. Here, we developed and screened highly diverse cyclic peptide libraries using mRNA display with a genetically encoded oxadiazolone (Ox) electrophile that was previously shown to potently and covalently inhibit multiple Fph enzymes. By performing multiple rounds of counter selections with WT and catalytic dead FphB, we were able to tune the selectivity of the resulting selected cyclic peptides containing the Ox residue toward the active site serine. From our mRNA display hits, we developed potent and selective fluorescent probes that label the active site of FphB at single digit nanomolar concentrations in live S. aureus bacteria. Taken together, this work demonstrates the potential of using direct genetically encoded electrophiles for mRNA display of covalent binding ligands and identifies potent new probes for FphB that have the potential to be used for diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Sijie Wang
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Emily C Woods
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Jeyun Jo
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Jiyun Zhu
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Althea Hansel-Harris
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, California 92037, United States
| | - Matthew Holcomb
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, California 92037, United States
| | - Manuel Llanos
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, California 92037, United States
| | - Nichole J Pedowitz
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Tulsi Upadhyay
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - John Bennett
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Matthias Fellner
- Biochemistry Department, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Ki Wan Park
- Department of Otolaryngology-Head & Neck Surgery Divisions, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Anna Zhang
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Tulio A Valdez
- Department of Otolaryngology-Head & Neck Surgery Divisions, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Stefano Forli
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, California 92037, United States
| | - Alix I Chan
- Department of Peptide Therapeutics, Genentech, South San Francisco, California 94080, United States
| | - Christian N Cunningham
- Department of Peptide Therapeutics, Genentech, South San Francisco, California 94080, United States
| | - Matthew Bogyo
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, United States
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, United States
| |
Collapse
|
22
|
Tao H, Yang B, Farhangian A, Xu K, Li T, Zhang ZY, Li J. Covalent-Allosteric Inhibitors: Do We Get the Best of Both Worlds? J Med Chem 2025; 68:4040-4052. [PMID: 39937154 DOI: 10.1021/acs.jmedchem.4c02760] [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: 02/13/2025]
Abstract
Covalent-allosteric inhibitors (CAIs) may achieve the best of both worlds: increased potency, long-lasting effects, and reduced drug resistance typical of covalent ligands, along with enhanced specificity and decreased toxicity inherent in allosteric modulators. Therefore, CAIs can be an effective strategy to transform many undruggable targets into druggable ones. However, CAIs are challenging to design. In this perspective, we analyze the discovery of known CAIs targeting three protein families: protein phosphatases, protein kinases, and GTPases. We also discuss how computational methods and tools can play a role in addressing the practical challenges of rational CAI design.
Collapse
Affiliation(s)
- Hui Tao
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Bo Yang
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Atena Farhangian
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ke Xu
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Tongtong Li
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Zhong-Yin Zhang
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Jianing Li
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
23
|
Aknine N, Pelletier R, Klymchenko AS. Lipid-Directed Covalent Labeling of Plasma Membranes for Long-Term Imaging, Barcoding and Manipulation of Cells. JACS AU 2025; 5:922-936. [PMID: 40017781 PMCID: PMC11863151 DOI: 10.1021/jacsau.4c01134] [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: 11/24/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 03/01/2025]
Abstract
Fluorescent probes for cell plasma membranes (PM) generally exploit a noncovalent labeling mechanism, which constitutes a fundamental limitation in multiple bioimaging applications. Here, we report a concept of lipid-directed covalent labeling of PM, which exploits transient binding to the lipid membrane surface generating a high local dye concentration, thus favoring covalent ligation to random proximal membrane proteins. This concept yielded fluorescent probes for PM called MemGraft, which are built of a dye (cyanine Cy3 or Cy5) bearing a low-affinity membrane anchor and a reactive group: an activated ester or a maleimide. In contrast to specially designed control dyes and commercial Cy3-based labels of amino or thiol groups, MemGraft probes stain efficiently PM, revealing the crucial role of the membrane anchor combined with optimal reactivity of the activated ester or the maleimide. MemGraft probes overcome existing limitations of noncovalent probes, which makes them compatible with cell fixation, permeabilization, trypsinization, and the presence of serum. The latter allows long-term cell tracking and video imaging of cell PM dynamics without the signs of phototoxicity. The covalent strategy also enables staining and long-term tracking of cocultured cells labeled in different colors without exchange of probes. Moreover, the combination of MemGraft-Cy3 and MemGraft-Cy5 probes at different ratios enabled long-term cell barcoding in at least 5 color codes, important for tracking and visualizing multiple populations of cells. Ultimately, we found that the MemGraft strategy enables efficient biotinylation of the cell surface, opening the path to cell surface engineering and cell manipulation.
Collapse
Affiliation(s)
- Nathan Aknine
- Laboratoire de Bioimagerie
et Pathologies, UMR 7021 CNRS, ITI SysChem-Chimie des Systèmes
Complexes, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Remi Pelletier
- Laboratoire de Bioimagerie
et Pathologies, UMR 7021 CNRS, ITI SysChem-Chimie des Systèmes
Complexes, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Andrey S. Klymchenko
- Laboratoire de Bioimagerie
et Pathologies, UMR 7021 CNRS, ITI SysChem-Chimie des Systèmes
Complexes, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| |
Collapse
|
24
|
Li H, Liu Y, Zhang S, Ma L, Zeng Z, Zhou Z, Gandon V, Xu H, Yi W, Wang S. Access to N-α-deuterated amino acids and DNA conjugates via Ca(II)-HFIP-mediated reductive deutero-amination of α-oxo-carbonyl compounds. Nat Commun 2025; 16:1816. [PMID: 39979333 PMCID: PMC11842556 DOI: 10.1038/s41467-025-57098-w] [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: 08/20/2024] [Accepted: 02/11/2025] [Indexed: 02/22/2025] Open
Abstract
The development of practical and selective strategies for deuterium incorporation to construct deuterated molecules, particularly deuterium-labeled amino acids, has become as a growing focus of basic research, yet it remains a formidable challenge. Herein, we present a bioinspired calcium-HFIP-mediated site-selective reductive deutero-amination of α-oxo-carbonyl compounds with amines. Utilizing d2-Hantzsch ester as the deuterium source, this reaction attains remarkable deuteration efficiency (> 99% deuteration). It enables the synthesis of N-α-deuterated amino acid motifs with a wide range of functionality, as evidenced by over 130 examples. The method exhibits compatibility with diverse substrates, such as amino acids, peptides, drug molecules, and natural products bearing different substituents. Moreover, the application of this strategy in the synthesis of DNA-tagged N-α-deuterated amino acids/peptides has been demonstrated. This work offers an efficient and innovative solution for deuterated amino acid chemistry and holds substantial application potential in organic synthesis, medicinal chemistry, and chemical biology.
Collapse
Affiliation(s)
- Haoran Li
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuwei Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Silin Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lei Ma
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhongyi Zeng
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhi Zhou
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Vincent Gandon
- Institut de Chimie Moléculaire et des Matériaux d'Orsay, CNRS UMR 8182, Université Paris-Saclay, Bâtiment Henri Moissan, Orsay, France
| | - Hongtao Xu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China.
| | - Wei Yi
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Shengdong Wang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
25
|
Kyoya T, Ishida H, Saitoh T, Itoh T. Preparation of ethynylsulfonamides and study of their reactivity with nucleophilic amino acids. Org Biomol Chem 2025; 23:1901-1908. [PMID: 39815739 DOI: 10.1039/d4ob01873g] [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: 01/18/2025]
Abstract
The development of covalent drugs, particularly those utilizing Michael acceptors, has garnered significant attention in recent pharmaceutical research due to the ability of such molecules to irreversibly inhibit protein function. This study focusses on the synthesis and evaluation of ethynylsulfonamides, which are predicted to have superior covalent binding ability, metabolic stability, and water solubility compared to traditional amides. We developed a straightforward synthesis method for ethynylsulfonamides and comprehensively evaluated the covalent binding abilities of these compounds using NMR with various nucleophilic amino acids in different solvents. Our results revealed that ethynylsulfonamides exhibit rapid and selective reactivity with cysteine residues, particularly in phosphate-buffered saline (PBS), where the reaction progressed quantitatively within five minutes. Notably, propynylsulfonamide demonstrated high reactivity and selectivity toward cysteine, suggestive of the significant potential of this molecule for applications in antibody-drug conjugates (ADCs) and other therapeutic areas where metabolic stability, water solubility, and selective reactivity are crucial.
Collapse
Affiliation(s)
- Tatsuhiro Kyoya
- Laboratory of Drug Design and Medicinal Chemistry, Showa Pharmaceutical University, 3-2-1 Higashi-Tamagawagakuen, Machida, Tokyo 194-8543, Japan.
| | - Hiroaki Ishida
- Laboratory of Drug Design and Medicinal Chemistry, Showa Pharmaceutical University, 3-2-1 Higashi-Tamagawagakuen, Machida, Tokyo 194-8543, Japan.
| | - Toshiaki Saitoh
- Division of Medicinal Informatics, Nihon Pharmaceutical University, 10281 Komuro, lna-machi, Kitaadachi-gun, Saitama 362-0806, Japan
| | - Toshimasa Itoh
- Laboratory of Drug Design and Medicinal Chemistry, Showa Pharmaceutical University, 3-2-1 Higashi-Tamagawagakuen, Machida, Tokyo 194-8543, Japan.
| |
Collapse
|
26
|
Hossain MA, Brahme RR, Miller BC, Amin J, de Barros M, Schneider JL, Auclair JR, Mattos C, Wang Q, Agar NYR, Greenblatt DJ, Manetsch R, Agar JN. Mass spectrometry methods and mathematical PK/PD model for decision tree-guided covalent drug development. Nat Commun 2025; 16:1777. [PMID: 39971904 PMCID: PMC11839910 DOI: 10.1038/s41467-025-56985-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/07/2025] [Indexed: 02/21/2025] Open
Abstract
Covalent drug discovery efforts are growing rapidly but have major unaddressed limitations. These include high false positive rates during hit-to-lead identification; the inherent uncoupling of covalent drug concentration and effect [i.e., uncoupling of pharmacokinetics (PK) and pharmacodynamics (PD)]; and a lack of bioanalytical and modeling methods for determining PK and PD parameters. We present a covalent drug discovery workflow that addresses these limitations. Our bioanalytical methods are based upon a mass spectrometry (MS) assay that can measure the percentage of drug-target protein conjugation (% target engagement) in biological matrices. Further we develop an intact protein PK/PD model (iPK/PD) that outputs PK parameters (absorption and distribution) as well as PD parameters (mechanism of action, protein metabolic half-lives, dose, regimen, effect) based on time-dependent target engagement data. Notably, the iPK/PD model is applicable to any measurement (e.g., bottom-up MS and other drug binding studies) that yields % of target engaged. A Decision Tree is presented to guide researchers through the covalent drug development process. Our bioanalytical methods and the Decision Tree are applied to two approved drugs (ibrutinib and sotorasib); the most common plasma off-target, human serum albumin; three protein targets (KRAS, BTK, SOD1), and to a promising SOD1-targeting ALS drug candidates.
Collapse
Affiliation(s)
- Md Amin Hossain
- Department of Chemistry and Chemical Biology, Northeastern University;Boston, Massachusetts, 02115, USA
- Barnett Institute of Chemical and Biological Analysis;Boston, Massachusetts, 02115, USA
- Department of Neurosurgery and Radiology, Brigham and Women's Hospital, Harvard Medical School;Boston, Massachusetts, 02115, USA
| | - Rutali R Brahme
- Department of Chemistry and Chemical Biology, Northeastern University;Boston, Massachusetts, 02115, USA
- Barnett Institute of Chemical and Biological Analysis;Boston, Massachusetts, 02115, USA
| | - Brandon C Miller
- Department of Chemistry and Chemical Biology, Northeastern University;Boston, Massachusetts, 02115, USA
| | - Jakal Amin
- Department of Chemistry and Chemical Biology, Northeastern University;Boston, Massachusetts, 02115, USA
- Barnett Institute of Chemical and Biological Analysis;Boston, Massachusetts, 02115, USA
| | - Marcela de Barros
- Department of Chemistry and Chemical Biology, Northeastern University;Boston, Massachusetts, 02115, USA
| | - Jaime L Schneider
- Massachusetts General Hospital Cancer Center, Harvard Medical School;Boston, Massachusetts, 02114, USA
| | - Jared R Auclair
- Department of Chemistry and Chemical Biology, Northeastern University;Boston, Massachusetts, 02115, USA
- Barnett Institute of Chemical and Biological Analysis;Boston, Massachusetts, 02115, USA
| | - Carla Mattos
- Department of Chemistry and Chemical Biology, Northeastern University;Boston, Massachusetts, 02115, USA
| | - Qingping Wang
- Sanofi US, Drug Metabolism and Pharmacokinetics;Cambridge, Massachusetts, 02141, USA
| | - Nathalie Y R Agar
- Department of Neurosurgery and Radiology, Brigham and Women's Hospital, Harvard Medical School;Boston, Massachusetts, 02115, USA
| | | | - Roman Manetsch
- Department of Chemistry and Chemical Biology, Northeastern University;Boston, Massachusetts, 02115, USA
- Department of Pharmaceutical Sciences, Northeastern University;Boston, Massachusetts, 02115, USA
| | - Jeffrey N Agar
- Department of Chemistry and Chemical Biology, Northeastern University;Boston, Massachusetts, 02115, USA.
- Barnett Institute of Chemical and Biological Analysis;Boston, Massachusetts, 02115, USA.
- Department of Pharmaceutical Sciences, Northeastern University;Boston, Massachusetts, 02115, USA.
| |
Collapse
|
27
|
Wang G, Seidler NJ, Röhm S, Pan Y, Liang XJ, Haarer L, Berger BT, Sivashanmugam SA, Wydra VR, Forster M, Laufer SA, Chaikuad A, Gehringer M, Knapp S. Probing the Protein Kinases' Cysteinome by Covalent Fragments. Angew Chem Int Ed Engl 2025; 64:e202419736. [PMID: 39716901 DOI: 10.1002/anie.202419736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 11/23/2024] [Accepted: 12/09/2024] [Indexed: 12/25/2024]
Abstract
Protein kinases are important drug targets, yet specific inhibitors have been developed for only a fraction of the more than 500 human kinases. A major challenge in designing inhibitors for highly related kinases is selectivity. Unlike their non-covalent counterparts, covalent inhibitors offer the advantage of selectively targeting structurally similar kinases by modifying specific protein side chains, particularly non-conserved cysteines. Previously, covalent fragment screens yielded potent and selective inhibitors for individual kinases such as ERK1/2 but have not been applied to the broader kinome. Furthermore, many of the accessible cysteine positions have not been addressed so far. Here, we outline a generalizable approach to sample ATP-site cysteines with fragment-like covalent inhibitors. We present the development of a kinase-focused covalent fragment library and its systematic screening against a curated selection of 47 kinases, with 60 active site-proximal cysteines using LC/MS and differential scanning fluorimetry (DSF) assays, followed by hit validation through various complementary techniques. Our findings expand the repertoire of targetable cysteines within protein kinases, provide insight into unique binding modes identified from crystal structures and deliver isoform-specific hits with promising profiles as starting points for the development of highly potent and selective covalent inhibitors.
Collapse
Affiliation(s)
- Guiqun Wang
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), DKTK Site Frankfurt-Mainz, German Cancer Research Center (DKFZ), D-69120, Heidelberg, Germany
| | - Nico J Seidler
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
| | - Sandra Röhm
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - Yufeng Pan
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - Xiaojun Julia Liang
- Faculty of Medicine, Institute for Biomedical Engineering, Department for Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided & Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Lisa Haarer
- Faculty of Medicine, Institute for Biomedical Engineering, Department for Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided & Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Benedict-Tilman Berger
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - Saran Aswathaman Sivashanmugam
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - Valentin R Wydra
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
| | - Michael Forster
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided & Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Stefan A Laufer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided & Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- Tübingen Center for Academic Drug Discovery & Development (TüCAD2), Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Apirat Chaikuad
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - Matthias Gehringer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
- Faculty of Medicine, Institute for Biomedical Engineering, Department for Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided & Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Stefan Knapp
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), DKTK Site Frankfurt-Mainz, German Cancer Research Center (DKFZ), D-69120, Heidelberg, Germany
| |
Collapse
|
28
|
Varone E, Retini M, Cherubini A, Chernorudskiy A, Marrazza A, Guidarelli A, Cagnotto A, Beeg M, Gobbi M, Fumagalli S, Bolis M, Guarrera L, Barbera MC, Grasselli C, Bleve A, Generali D, Milani M, Mari M, Salmona M, Piersanti G, Bottegoni G, Broggini M, Janssen-Heininger YMW, Cho J, Cantoni O, Zito E. Small molecule-mediated inhibition of the oxidoreductase ERO1A restrains aggressive breast cancer by impairing VEGF and PD-L1 in the tumor microenvironment. Cell Death Dis 2025; 16:105. [PMID: 39962052 PMCID: PMC11833095 DOI: 10.1038/s41419-025-07426-1] [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: 09/18/2024] [Revised: 01/09/2025] [Accepted: 02/03/2025] [Indexed: 02/20/2025]
Abstract
Cancer cells adapt to harsh environmental conditions by inducing the Unfolded Protein Response (UPR), of which ERO1A is a mediator. ERO1A aids protein folding by acting as a protein disulfide oxidase, and under cancer-related hypoxia conditions, it favors the folding of angiogenic VEGFA, leading tumor cells to thrive and spread. The upregulation of ERO1A in cancer cells, oppositely to the dispensability of ERO1A activity in healthy cells, renders ERO1A a perfect target for cancer therapy. Here, we report the upregulation of ERO1A in a cohort of aggressive triple-negative breast cancer (TNBC) patients in which ERO1A levels correlate with a higher risk of breast tumor recurrence and metastatic spread. For ERO1A target validation and therapy in TNBC, we designed new ERO1A inhibitors in a structure-activity campaign of the prototype EN460. Cell-based screenings showed that the presence of the Micheal acceptor in the compound is necessary to engage the cysteine 397 of ERO1A but not sufficient to set out the inhibitory effect on ERO1A. Indeed, the ERO1 inhibitor must adopt a non-coplanar rearrangement within the ERO1A binding site. I2 and I3, two new EN460 analogs with different phenyl-substituted moieties, efficiently inhibited ERO1A, blunting VEGFA secretion. Accordingly, in vitro assays to measure ERO1A engagement and inhibition confirmed that I2 and I3 bind ERO1A and restrain its activity with a IC50 in a low micromolar range. EN460, I2 and I3 triggered breast cancer cytotoxicity while specifically inhibiting ERO1A in a dose-dependent manner. I2 more efficiently impaired cancer-relevant features such as VEGFA secretion and related cell migration. I2 also acted on the tumor microenvironment and viability of xenografts and syngeneic TNBC. Thus, small molecule-mediated ERO1A pharmacological inhibition is feasible and promises to lead to effective therapy for the still incurable TNBC.
Collapse
Affiliation(s)
- Ersilia Varone
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Michele Retini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Alessandro Cherubini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Alexander Chernorudskiy
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- School of Medicine, Taizhou University, Taizhou, 318000, Zhejiang, China
| | - Alice Marrazza
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Andrea Guidarelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Alfredo Cagnotto
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marten Beeg
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marco Gobbi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Marco Bolis
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Bioinformatics Core Unit, Institute of Oncology Research (IOR), Bellinzona, Switzerland
| | - Luca Guarrera
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Chiara Grasselli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Augusto Bleve
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Daniele Generali
- U.O. Patologia Mammaria e Tumori Cerebrali, Azienda Socio-Sanitaria Territoriale, Cremona, Italia
| | - Manuela Milani
- U.O. Patologia Mammaria e Tumori Cerebrali, Azienda Socio-Sanitaria Territoriale, Cremona, Italia
| | - Michele Mari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Mario Salmona
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Giovanni Piersanti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Giovanni Bottegoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
- Institute of Clinical Sciences, University of Birmingham, Edgbaston, B15 2TT, Birmingham, UK
| | - Massimo Broggini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Yvonne M W Janssen-Heininger
- Departments of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, USA
| | - Orazio Cantoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| |
Collapse
|
29
|
Zammit CM, Nadel CM, Lin Y, Koirala S, Potts PR, Nomura DK. Covalent Destabilizing Degrader of AR and AR-V7 in Androgen-Independent Prostate Cancer Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637117. [PMID: 39990458 PMCID: PMC11844536 DOI: 10.1101/2025.02.12.637117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Androgen-independent prostate cancers, correlated with heightened aggressiveness and poor prognosis, are caused by mutations or deletions in the androgen receptor (AR) or expression of truncated variants of AR that are constitutively activated. Currently, drugs and drug candidates against AR target the steroid-binding domain to antagonize or degrade AR. However, these compounds cannot therapeutically access largely intrinsically disordered truncated splice variants of AR, such as AR-V7, that only possess the DNA binding domain and are missing the ligand binding domain. Targeting intrinsically disordered regions within transcription factors has remained challenging and is considered "undruggable". Herein, we leveraged a cysteine-reactive covalent ligand library in a cellular screen to identify degraders of AR and AR-V7 in androgen-independent prostate cancer cells. We identified a covalent compound EN1441 that selectively degrades AR and AR-V7 in a proteasome-dependent manner through direct covalent targeting of an intrinsically disordered cysteine C125 in AR and AR-V7. EN1441 causes significant and selective destabilization of AR and AR-V7, leading to aggregation of AR/AR-V7 and subsequent proteasome-mediated degradation. Consistent with targeting both AR and AR-V7, we find that EN1441 completely inhibits total AR transcriptional activity in androgen-independent prostate cancer cells expressing both AR and AR-V7 compared to AR antagonists or degraders that only target the ligand binding domain of full-length AR, such as enzalutamide and ARV-110. Our results put forth a pathfinder molecule EN1441 that targets an intrinsically disordered cysteine within AR to destabilize, degrade, and inhibit both AR and AR-V7 in androgen-independent prostate cancer cells and highlights the utility of covalent ligand discovery approaches in directly targeting, destabilizing, inhibiting, and degrading classically undruggable transcription factor targets.
Collapse
Affiliation(s)
- Charlotte M. Zammit
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720 USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720 USA
- Innovative Genomics Institute, Berkeley, CA 94720 USA
| | - Cory M. Nadel
- Induced Proximity Platform, Amgen Research, Thousand Oaks, CA 91320 USA
| | - Ying Lin
- Induced Proximity Platform, Amgen Research, Thousand Oaks, CA 91320 USA
| | - Sajjan Koirala
- Induced Proximity Platform, Amgen Research, Thousand Oaks, CA 91320 USA
| | | | - Daniel K. Nomura
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720 USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720 USA
- Innovative Genomics Institute, Berkeley, CA 94720 USA
| |
Collapse
|
30
|
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 2025; 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] [MESH Headings] [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
|
31
|
Dalton SE, Di Pietro O, Hennessy E. A Medicinal Chemistry Perspective on FDA-Approved Small Molecule Drugs with a Covalent Mechanism of Action. J Med Chem 2025; 68:2307-2313. [PMID: 39899741 DOI: 10.1021/acs.jmedchem.4c02661] [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: 02/05/2025]
Abstract
Covalent modification of disease-driving proteins as a therapeutic strategy has experienced a well-documented resurgence since 2010. However, the earliest FDA approval dates for covalent drugs are in the 1940s, although the covalent mechanism of action may not have been known at the time. This article discloses a data set of all FDA-approved small molecule drugs acting via a covalent mechanism of action, annotated by indication, biological target, reactive group on the drug, biological reactive partner (i.e., amino acid residue, cofactor, etc.), chemical reaction mechanism, bioactivation requirements, key references, and reversibility profile. We discuss these data in the context of addressing key questions posed by the Merck Discovery Chemistry community when considering a chemical series with a covalent mechanism of action.
Collapse
Affiliation(s)
- Samuel E Dalton
- Department of Discovery Chemistry, MSD, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom
| | - Ornella Di Pietro
- Department of Discovery Chemistry, MSD, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom
| | - Elisabeth Hennessy
- Department of Discovery Chemistry, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| |
Collapse
|
32
|
Zhang Z, Gao R, Zhao M, Zhang X, Gao H, Qi Y, Wang R, Li Y. Computational Methods for Predicting Chemical Reactivity of Covalent Compounds. J Chem Inf Model 2025; 65:1140-1154. [PMID: 39823568 DOI: 10.1021/acs.jcim.4c01591] [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: 01/19/2025]
Abstract
In recent decades, covalent inhibitors have emerged as a promising strategy for therapeutic development, leveraging their unique mechanism of forming covalent bonds with target proteins. This approach offers advantages such as prolonged drug efficacy, precise targeting, and the potential to overcome resistance. However, the inherent reactivity of covalent compounds presents significant challenges, leading to off-target effects and toxicities. Accurately predicting and modulating this reactivity have become a critical focus in the field. In this work, we compiled a data set of 419 cysteine-targeted covalent compounds and their reactivity through an extensive literature review. Employing machine learning, deep learning, and quantum mechanical calculations, we evaluated the intrinsic reactivity of the covalent compounds. Our FP-Stack models demonstrated robust Pearson and Spearman correlations of approximately 0.80 and 0.75 on the test set, respectively. This empowers rapid and accurate reactivity predictions, significantly reducing computational costs and streamlining structural handling and experimental procedures. Experimental validation on acrylamide compounds underscored the predictive efficacy of our model. This study presents an efficient computational tool for the reactivity prediction of covalent compounds and is expected to offer valuable insights for guiding covalent drug discovery and development.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| | - Ruyu Gao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| | - Meiling Zhao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| | - Xiangying Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| | - Haotian Gao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| | - Yifei Qi
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| | - Renxiao Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| | - Yan Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| |
Collapse
|
33
|
Wang S, Zhong Z. Integrating targeting and drug retention: A promising approach for cancer treatment. J Control Release 2025; 378:247-249. [PMID: 39667567 DOI: 10.1016/j.jconrel.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Affiliation(s)
- Shenqiang Wang
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China.
| | - Zhiyuan Zhong
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, PR China; International College of Pharmaceutical Innovation, Soochow University, Suzhou 215200, PR China.
| |
Collapse
|
34
|
Edwards AN, Hsu KL. Emerging opportunities for intact and native protein analysis using chemical proteomics. Anal Chim Acta 2025; 1338:343551. [PMID: 39832869 DOI: 10.1016/j.aca.2024.343551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025]
Abstract
Chemical proteomics has advanced small molecule ligand discovery by providing insights into protein-ligand binding mechanism and enabling medicinal chemistry optimization of protein selectivity on a global scale. Mass spectrometry is the predominant analytical method for chemoproteomics, and various approaches have been deployed to investigate and target a rapidly growing number of protein classes and biological systems. Two methods, intact mass analysis (IMA) and top-down proteomics (TDMS), have gained interest in recent years due to advancements in high resolution mass spectrometry instrumentation. Both methods apply mass spectrometry analysis at the proteoform level, as opposed to the peptide level of bottom-up proteomics (BUMS), thus addressing some of the challenges of protein inference and incomplete information on modification stoichiometry. This Review covers recent research progress utilizing MS-based proteomics methods, discussing in detail the capabilities and opportunities for improvement of each method. Further, heightened attention is given to IMA and TDMS, highlighting these methods' strengths and considerations when utilized in chemoproteomic studies. Finally, we discuss the capabilities of native mass spectrometry (nMS) and ion mobility mass spectrometry (IM-MS) and how these methods can be used in chemoproteomics research to complement existing approaches to further advance the field of functional proteomics.
Collapse
Affiliation(s)
- Alexis N Edwards
- Department of Chemistry, University of Texas at Austin, Austin, TX, 78712, United States
| | - Ku-Lung Hsu
- Department of Chemistry, University of Texas at Austin, Austin, TX, 78712, United States.
| |
Collapse
|
35
|
Lei Q, Zhou X, Li Y, Zhao S, Yang N, Xiao Z, Song C, Yu Q, Deng H. Image-Based Phenotypic Profiling Enables Rapid and Accurate Assessment of EGFR-Activating Mutations in Tissues from Lung Cancer Patients. J Am Chem Soc 2025; 147:4552-4570. [PMID: 39745025 DOI: 10.1021/jacs.4c16528] [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: 02/06/2025]
Abstract
Determining mutations in the kinase domain of the epidermal growth factor receptor (EGFR) is critical for the effectiveness of EGFR tyrosine kinase inhibitors (TKIs) in lung cancer. Yet, DNA-based sequencing analysis of tumor samples is time-consuming and only provides gene mutation information on EGFR, making it challenging to design effective EGFR-TKI therapeutic strategies. Here, we present a new image-based method involving the rational design of a quenched probe based on EGFR-TKI to identify mutant proteins, which permits specific and "no-wash" real-time imaging of EGFR in living cells only upon covalent targeting of the EGFR kinase. We also show that the probe enables distinguishing EGFR mutant tumor-bearing mice from wild-type tumor-bearing mice via fluorescence-intensity-based imaging with high signal contrast. More interestingly, the image-based phenotypic approach can be used to predict EGFR mutations in tumors from lung cancer patients with an accuracy of 94%. Notably, when immunohistochemistry analysis is integrated, an improved accuracy of 98% is achieved. These data delineate a drug-based phenotypic imaging approach for in-biopsy visualization and define functional groups of EGFR mutants that can effectively guide EGFR-TKI therapeutic decision-making besides gene mutation analysis.
Collapse
Affiliation(s)
- Qian Lei
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610065, China
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610065, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Xinglong Zhou
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China
| | - Ying Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610065, China
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610065, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Shuang Zhao
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Na Yang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Zhaolin Xiao
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610065, China
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610065, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Chao Song
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610065, China
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610065, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Quanwei Yu
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Hui Deng
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610065, China
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610065, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610065, China
| |
Collapse
|
36
|
Xiao Z, Gavriil ES, Cao F, Zhang X, Li SX, Kotelnikov S, Michalska P, Marte F, Huang C, Lu Y, Zhang Y, Bernardini E, Kozakov D, Tate EW. Identification of actionable targeted protein degradation effector sites through Site-specific Ligand Incorporation-induced Proximity (SLIP). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.04.636303. [PMID: 39975383 PMCID: PMC11838594 DOI: 10.1101/2025.02.04.636303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Targeted protein degradation (TPD) is a rapidly emerging and potentially transformative therapeutic modality. However, the large majority of >600 known ubiquitin ligases have yet to be exploited as TPD effectors by proteolysis-targeting chimeras (PROTACs) or molecular glue degraders (MGDs). We report here a chemical-genetic platform, Site-specific Ligand Incorporation-induced Proximity (SLIP), to identify actionable ("PROTACable") sites on any potential effector protein in intact cells. SLIP uses genetic code expansion (GCE) to encode copper-free "click" ligation at a specific effector site in intact cells, enabling in situ formation of a covalent PROTAC-effector conjugate against a target protein of interest (POI). Modification at actionable effector sites drives degradation of the targeted protein, establishing the potential of these sites for TPD. Using SLIP, we systematically screened dozens of sites across E3 ligases and E2 enzymes from diverse classes, identifying multiple novel potentially PROTACable effector sites which are competent for TPD. SLIP adds a powerful approach to the proximity-induced pharmacology (PIP) toolbox, enabling future effector ligand discovery to fully enable TPD, and other emerging PIP modalities.
Collapse
Affiliation(s)
- Zhangping Xiao
- Department of Chemistry, Imperial College London, 82 Wood Lane, London W12 0BZ, UK
| | | | - Fangyuan Cao
- Department of Chemistry, Imperial College London, 82 Wood Lane, London W12 0BZ, UK
| | - Xinyue Zhang
- Department of Chemistry, Imperial College London, 82 Wood Lane, London W12 0BZ, UK
| | - Stan Xiaogang Li
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794, United States
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, United States
| | - Sergei Kotelnikov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794, United States
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, United States
| | - Patrycja Michalska
- Department of Chemistry, Imperial College London, 82 Wood Lane, London W12 0BZ, UK
| | - Friederike Marte
- Department of Chemistry, Imperial College London, 82 Wood Lane, London W12 0BZ, UK
| | - Chloe Huang
- Department of Chemistry, Imperial College London, 82 Wood Lane, London W12 0BZ, UK
| | - Yudi Lu
- Department of Chemistry, Imperial College London, 82 Wood Lane, London W12 0BZ, UK
| | - Yunxuan Zhang
- Department of Chemistry, Imperial College London, 82 Wood Lane, London W12 0BZ, UK
| | - Erika Bernardini
- Department of Chemistry, Imperial College London, 82 Wood Lane, London W12 0BZ, UK
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794, United States
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, United States
| | - Edward W. Tate
- Department of Chemistry, Imperial College London, 82 Wood Lane, London W12 0BZ, UK
| |
Collapse
|
37
|
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 2025; 64:e202418314. [PMID: 39630105 DOI: 10.1002/anie.202418314] [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: 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
|
38
|
Zhou M, Li S, Tan Y, Huang W, Li Y, Yuan X, Li Z. Global Profiling Lysine Reactivity and Ligandability with Oxidant-Triggered Bioconjugation Chemistry. Angew Chem Int Ed Engl 2025; 64:e202418473. [PMID: 39543955 DOI: 10.1002/anie.202418473] [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: 09/25/2024] [Revised: 11/14/2024] [Accepted: 11/14/2024] [Indexed: 11/17/2024]
Abstract
Due to the high abundance and diverse functions of lysine residues, both in the interior and on the surface of proteins, the development of new methods to characterize their reactivity and ligandability could significantly expand the pool of druggable targets. To date, only a limited number of aminophilic electrophiles have been assessed for interactions with the lysine proteome, resulting in a substantial fraction remaining inaccessible to current probes. Here, to the best of our knowledge, we report the first oxidant-triggered bioconjugation platform for in-depth profiling of lysines. We quantified over 7000 covalently modifiable lysine residues, which significantly expands the coverage of ligandable lysines in the whole proteome. Chemical proteomics enabled the mapping of more than 100 endogenous kinases, thus providing a comprehensive landscape of ligandable catalytic lysines within the kinome. Moreover, we identified a suite of new ligandable lysines such as K60 of ENO1 and K31 of PPIA, offering insights for exploring new functional and targetable residues. These findings could provide valuable clues for the development of targeted covalent inhibitors (TCIs).
Collapse
Affiliation(s)
- Mengya Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development (MOE), School of Pharmacy, Jinan University, 601 Huangpu Avenue West, 510632, Guangzhou, China
| | - Shengrong Li
- Guangdong Second Provincial General Hospital, Postdoctoral Station of Traditional Chinese Medicine, Jinan University, 510632, Guangzhou, China
| | - Yi Tan
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development (MOE), School of Pharmacy, Jinan University, 601 Huangpu Avenue West, 510632, Guangzhou, China
| | - Weizhen Huang
- The First Huizhou Affiliated Hospital of Guangdong Medical University, 516001, Huizhou, China
| | - Yifang Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development (MOE), School of Pharmacy, Jinan University, 601 Huangpu Avenue West, 510632, Guangzhou, China
| | - Xia Yuan
- The First Huizhou Affiliated Hospital of Guangdong Medical University, 516001, Huizhou, China
| | - Zhengqiu Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development (MOE), School of Pharmacy, Jinan University, 601 Huangpu Avenue West, 510632, Guangzhou, China
| |
Collapse
|
39
|
Chodup P, Samodelov SL, Visentin M, Kullak‐Ublick GA. Drug-Induced Liver Injury Associated With Emerging Cancer Therapies. Liver Int 2025; 45:e70002. [PMID: 39853863 PMCID: PMC11760653 DOI: 10.1111/liv.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/26/2025]
Abstract
Targeted therapies and immunotherapies have shown great promise as best-in-class treatments for several cancers with respect to efficacy and safety. While liver test abnormalities are rather common in patients treated with kinase inhibitors or immunotherapy, events of severe hepatotoxicity in these patients are rare in comparison with those associated with chemotherapeutics. The underlying mechanisms and risk factors for severe hepatotoxicity with novel oncology therapies are not well understood, complicating the drug-induced liver injury (DILI) risk assessment in the preclinical and clinical phases of drug development. The epidemiological and clinical characteristics, as well as mechanisms of liver toxicity, are described here to the current state of knowledge. Tools to study and assess the risk of DILI during drug development are concisely summarised, focusing on caveats thereof for novel oncology treatments. Emerging tools to optimise safety assessments and gather additional mechanistic insights into DILI are introduced. Particularly in oncology, where standard liver signals during drug development are tolerated to a marginally higher degree than in other indications due to the life-saving, life-extending and quality-of-life improvements for patients with severe or advanced cancers versus previous standard-of-care therapeutics, safety assessments must be tailored to the drug and indication. Trends in patient safety-centred drug development programmes and regulatory approval processes must continually be revisited and streamlined via obtaining an overall greater understanding of DILI and the tools available to assess mechanisms of injury, frequency, severity and prognosis.
Collapse
Affiliation(s)
- Piotr Chodup
- Department of Clinical Pharmacology and ToxicologyUniversity Hospital Zürich, University of ZürichZürichSwitzerland
| | - Sophia L. Samodelov
- Department of Clinical Pharmacology and ToxicologyUniversity Hospital Zürich, University of ZürichZürichSwitzerland
| | - Michele Visentin
- Department of Clinical Pharmacology and ToxicologyUniversity Hospital Zürich, University of ZürichZürichSwitzerland
| | - Gerd A. Kullak‐Ublick
- Department of Clinical Pharmacology and ToxicologyUniversity Hospital Zürich, University of ZürichZürichSwitzerland
- Mechanistic Safety, Patient Safety & Pharmacovigilance, Novartis DevelopmentBaselSwitzerland
| |
Collapse
|
40
|
Rudalska R, Harbig J, Forster M, Woelffing P, Esposito A, Kudolo M, Botezatu A, Haller V, Janssen N, Holzmayer S, Nahidino P, Trompak O, Pantsar T, Kronenberger T, Yurttas C, Rist E, Weber ANR, Dahlke MH, Ott G, Koenigsrainer A, Rothbauer U, Maerklin M, Muerdter T, Schwab M, Singer S, Zender L, Laufer S, Dauch D. First-in-class ultralong-target-residence-time p38α inhibitors as a mitosis-targeted therapy for colorectal cancer. NATURE CANCER 2025; 6:259-277. [PMID: 39820127 DOI: 10.1038/s43018-024-00899-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 12/12/2024] [Indexed: 01/19/2025]
Abstract
Colorectal cancer (CRC) constitutes the second leading cause of cancer-related death worldwide and advanced CRCs are resistant to targeted therapies, chemotherapies and immunotherapies. p38α (Mapk14) has been suggested as a therapeutic target in CRC; however, available p38α inhibitors only allow for insufficient target inhibition. Here we describe a unique class of p38α inhibitors with ultralong target residence times (designated ULTR-p38i) that robustly inhibit p38α downstream signaling and induce distinct biological phenotypes. ULTR-p38i monotherapy triggers an uncontrolled mitotic entry by activating Cdc25 and simultaneously blocking Wee1. Consequently, CRC cells undergo mitotic catastrophe, resulting in apoptosis or senescence. ULTR-p38i exhibit high selectivity, good pharmaco-kinetic properties and no measurable toxicity with strong therapeutic effects in patient-derived CRC organoids and syngeneic CRC mouse models. Conceptually, our study suggests ultralong-target-residence-time kinase inhibitors as an alternative to covalent inhibitors, which, because of the lack of cysteine residues, cannot be generated for many kinase cancer targets.
Collapse
Affiliation(s)
- Ramona Rudalska
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Jule Harbig
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Michael Forster
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
| | - Pascal Woelffing
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Aylin Esposito
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Mark Kudolo
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
| | - Adelina Botezatu
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Vanessa Haller
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
| | - Nicole Janssen
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart and University of Tübingen, Tübingen, Germany
| | - Samuel Holzmayer
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Philipp Nahidino
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
| | - Omelyan Trompak
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Tatu Pantsar
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Thales Kronenberger
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Can Yurttas
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Elke Rist
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Alexander N R Weber
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Marc H Dahlke
- Department of General and Visceral Surgery, Robert Bosch Hospital, Stuttgart, Germany
| | - German Ott
- Department of Clinical Pathology, Robert Bosch Hospital, Stuttgart, Germany
| | - Alfred Koenigsrainer
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Ulrich Rothbauer
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
- Department of Pharmaceutical Biotechnology, University of Tübingen, Tübingen, Germany
| | - Melanie Maerklin
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Thomas Muerdter
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart and University of Tübingen, Tübingen, Germany
| | - Matthias Schwab
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart and University of Tübingen, Tübingen, Germany
- Departments of Clinical Pharmacology, and of Biochemistry and Pharmacy, University of Tübingen, Tübingen, Germany
- German Cancer Research Consortium (DKTK), Partner Site Tübingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephan Singer
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Institute of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Lars Zender
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- German Cancer Research Consortium (DKTK), Partner Site Tübingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Tübingen Center for Academic Drug Discovery and Development (TüCAD2), Tübingen, Germany
| | - Stefan Laufer
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
- Tübingen Center for Academic Drug Discovery and Development (TüCAD2), Tübingen, Germany
| | - Daniel Dauch
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany.
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany.
- Tübingen Center for Academic Drug Discovery and Development (TüCAD2), Tübingen, Germany.
| |
Collapse
|
41
|
Faridoon, Zheng J, Zhang G, Li JJ. Key advances in the development of reversible covalent inhibitors. Future Med Chem 2025; 17:389-392. [PMID: 39829174 PMCID: PMC11834458 DOI: 10.1080/17568919.2025.2453407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025] Open
Affiliation(s)
- Faridoon
- Center for Drug Design and Development, Suzhou Genhouse Bio Co., Ltd., Suzhou, Jiangsu, China
| | - Jiyue Zheng
- Center for Drug Design and Development, Suzhou Genhouse Bio Co., Ltd., Suzhou, Jiangsu, China
| | - Guiping Zhang
- Center for Drug Design and Development, Suzhou Genhouse Bio Co., Ltd., Suzhou, Jiangsu, China
| | - Jie Jack Li
- Center for Drug Design and Development, Suzhou Genhouse Bio Co., Ltd., Suzhou, Jiangsu, China
| |
Collapse
|
42
|
Sarker A, Uddin B, Ahmmed R, Mahmud S, Ajadee A, Pappu MAA, Aziz MA, Mollah MNH. Discovery of mutated oncodriver genes associated with glioblastoma originated from stem cells of subventricular zone through whole exome sequence profile analysis, and drug repurposing. Heliyon 2025; 11:e42052. [PMID: 39906820 PMCID: PMC11791140 DOI: 10.1016/j.heliyon.2025.e42052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 02/06/2025] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive cancers due to its high mortality rate in spite of intensive treatment. It may be happened because of drug resistance against their typical receptors, since these receptor genes are often mutated by environmental stress. So identifying mutated oncodriver genes which could be used as potential drug target is essential in order to develop effective new therapeutic drugs as well as better prognosis for GBM patients. In this study, we analyzed whole exome sequencing (WES) profiles of NCBI database on GBM and matched-normal (control) samples originated from astrocyte like neural stem cells (NSC) of subventricular zone (SVZ) to explore GBM-causing mutated oncodriver genes, since SVZ is considered as the origin of GBM development. We detected 16 mutated oncodriver genes. Then, filtering by differential co-expression analysis based on independent RNA-Seq profiles of CGGA database revealed 10 genes as dysregulated oncodriver genes. Following that, 3 significantly overexpressed oncodriver genes (MTCH2, VWF, and WDR89) were identified as potential drug targets. Then molecular mechanisms of GBM development were investigated by these three overexpressed driver genes through gene ontology (GO), KEGG-pathways, Gene regulatory network (GRN) and mutation analysis. Finally, overexpressed oncodriver genes guided top-ranked six drug agents (Irinotecan, Imatinib, etoposide, pazopanib, trametinib and cabozanitinib) were recommended against GBM through molecular docking study. Most of our findings received support by the literature review also. Therefore, the findings of this study might carry potential values to the wet-lab researchers for further investigation in terms of diagnosis and therapies of GBM.
Collapse
Affiliation(s)
- Arnob Sarker
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Burhan Uddin
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Reaz Ahmmed
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Sabkat Mahmud
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Alvira Ajadee
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Md. Al Amin Pappu
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Md. Abdul Aziz
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Md. Nurul Haque Mollah
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
| |
Collapse
|
43
|
Tan YS, Chakrabarti M, Stein RM, Prentis LE, Rizzo RC, Kurtzman T, Fischer M, Balius TE. Development of Receptor Desolvation Scoring and Covalent Sampling in DOCK 6: Methods Evaluated on a RAS Test Set. J Chem Inf Model 2025; 65:722-748. [PMID: 39757424 PMCID: PMC11776051 DOI: 10.1021/acs.jcim.4c01623] [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: 09/09/2024] [Revised: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 01/07/2025]
Abstract
Molecular docking methods are widely used in drug discovery efforts. RAS proteins are important cancer drug targets, and are useful systems for evaluating docking methods, including accounting for solvation effects and covalent small molecule binding. Water often plays a key role in small molecule binding to RAS proteins, and many inhibitors─including FDA-approved drugs─covalently bind to oncogenic RAS proteins. We assembled a RAS test set, consisting of 138 RAS protein structures and 2 structures of KRAS DNA in complex with ligands. In DOCK 6, we have implemented a receptor desolvation scoring function and a covalent docking algorithm. These new features were evaluated using the test set, with pose reproduction, cross-docking, and enrichment calculations. We tested two solvation methods for generating receptor desolvation scoring grids: GIST and 3D-RISM. Using grids from GIST or 3D-RISM, water displacements are precomputed with Gaussian-weighting, and trilinear interpolation is used to speed up this scoring calculation. To test receptor desolvation scoring, we prepared GIST and 3D-RISM grids for all KRAS systems in the test set, and we compare enrichment performance with and without receptor desolvation. Accounting for receptor desolvation using GIST improves enrichment for 51% of systems and worsens enrichment for 35% of systems, while using 3D-RISM improves enrichment for 44% of systems and worsens enrichment for 30% of systems. To more rigorously test accounting for receptor desolvation using 3D-RISM, we compare pose reproduction with and without 3D-RISM receptor desolvation. Pose reproduction docking with 3D-RISM yields a 1.8 ± 2.41% increase in success rate compared to docking without 3D-RISM. Accounting for receptor desolvation provides a small, but significant, improvement in both enrichment and pose reproduction for this set. We tested the covalent attach-and-grow algorithm on 70 KRAS systems containing covalent ligands, obtaining similar pose reproduction success rates between covalent and noncovalent docking. Comparing covalent docking to noncovalent docking, there is a 2.4 ± 3.29% increase and a 1.27 ± 3.33% decline in the success rate when docking with experimental and SMILES-generated ligand conformations, respectively. As a proof-of-concept, we performed covalent virtual screens with and without receptor desolvation scoring, targeting the switch II pocket of KRAS, using 3.4 million make-on-demand acrylamide compounds from the Enamine REAL database. On average, the attach-and-grow algorithm spends approximately 17.61 s per molecule across the screen. The test set is available at https://github.com/tbalius/teb_docking_test_sets.
Collapse
Affiliation(s)
- Y. Stanley Tan
- NCI
RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical
Research, Inc., P.O. Box B, Frederick 21702, Maryland, United States
| | - Mayukh Chakrabarti
- NCI
RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical
Research, Inc., P.O. Box B, Frederick 21702, Maryland, United States
| | - Reed M. Stein
- Department
of Pharmaceutical Chemistry, University
of California—San Francisco, San Francisco 94158, California, United States
| | - Lauren E. Prentis
- Department
of Biochemistry and Structural Biology, Stony Brook University, Stony
Brook 11794, New York, United States
- Institute
of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook11794, New York, United States
| | - Robert C. Rizzo
- Institute
of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook11794, New York, United States
- Department
of Applied Mathematics and Statistics, Stony
Brook University, Stony Brook 11794, New York, United States
- Laufer Center
for Physical and Quantitative Biology, Stony
Brook University, Stony Brook11794, New York, United States
| | - Tom Kurtzman
- PhD
Programs in Chemistry, Biochemistry, and Biology, The Graduate Center of the City University of New York, New York 10016, New York, United States
- Department
of Chemistry, Lehman College, The City University
of New York, Bronx 10468, New York, United States
| | - Marcus Fischer
- Department
of Chemical Biology and Therapeutics, St.
Jude Children’s Research Hospital, Memphis38105, Tennessee, United States
| | - Trent E. Balius
- NCI
RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical
Research, Inc., P.O. Box B, Frederick 21702, Maryland, United States
| |
Collapse
|
44
|
Reimer BM, Awoonor-Williams E, Golosov AA, Hornak V. CovCysPredictor: Predicting Selective Covalently Modifiable Cysteines Using Protein Structure and Interpretable Machine Learning. J Chem Inf Model 2025; 65:544-553. [PMID: 39780548 DOI: 10.1021/acs.jcim.4c01281] [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: 01/11/2025]
Abstract
Targeted covalent inhibition is a powerful therapeutic modality in the drug discoverer's toolbox. Recent advances in covalent drug discovery, in particular, targeting cysteines, have led to significant breakthroughs for traditionally challenging targets such as mutant KRAS, which is implicated in diverse human cancers. However, identifying cysteines for targeted covalent inhibition is a difficult task, as experimental and in silico tools have shown limited accuracy. Using the recently released CovPDB and CovBinderInPDB databases, we have trained and tested interpretable machine learning (ML) models to identify cysteines that are liable to be covalently modified (i.e., "ligandable" cysteines). We explored myriad physicochemical features (pKa, solvent exposure, residue electrostatics, etc.) and protein-ligand pocket descriptors in our ML models. Our final logistic regression model achieved a median F1 score of 0.73 on held-out test sets. When tested on a small sample of holo proteins, our model also showed reasonable performance, accurately predicting the most ligandable cysteine in most cases. Taken together, these results indicate that we can accurately predict potential ligandable cysteines for targeted covalent drug discovery, privileging cysteines that are more likely to be selective rather than purely reactive. We release this tool to the scientific community as CovCysPredictor.
Collapse
Affiliation(s)
- Bryn Marie Reimer
- Computer-Aided Drug Discovery, Global Discovery Chemistry, Novartis Biomedical Research, 181 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Manning College of Information & Computer Sciences, University of Massachusetts Amherst, 140 Governors Drive, Amherst, Massachusetts 01003, United States
| | - Ernest Awoonor-Williams
- Computer-Aided Drug Discovery, Global Discovery Chemistry, Novartis Biomedical Research, 181 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Andrei A Golosov
- Computer-Aided Drug Discovery, Global Discovery Chemistry, Novartis Biomedical Research, 181 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Viktor Hornak
- Computer-Aided Drug Discovery, Global Discovery Chemistry, Novartis Biomedical Research, 181 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
45
|
Zhong Z, Hocking BJW, Brown CP, Ma TK, White AJP, Mann DJ, Armstrong A, Bull JA. Synthesis and Functionalization of Sulfoximine-Bicyclo[1.1.0]butanes: Functionalizable, Tuneable and Cysteine-Selective Chiral Warheads. Angew Chem Int Ed Engl 2025; 64:e202420028. [PMID: 39551713 DOI: 10.1002/anie.202420028] [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: 10/16/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 11/19/2024]
Abstract
Electrophilic covalent warheads with appropriate reactivity and selectivity are crucial to the investigation of protein function and the discovery of therapeutics. Here we report the synthesis of sulfoximine bicyclo[1.1.0] butanes (BCBs) as novel thiol reactive chiral warheads, achieved in one-pot from methylsulfoximines. Unusually the warhead can then be derivatized, keeping the BCB intact, over 3 vectors: i) sulfoximine N-modification instills a broad range of strain-release reactivity; ii) sp2-cross-coupling reactions on aryl-BCB-sulfoximines allows direct diversification, and iii) functionalization of the BCB motif itself is achieved by metalation and trapping with electrophiles. The BCB sulfoximines are shown to react selectively with cysteine including in a protein model (CDK2) under biocompatible conditions. Preliminary data indicate suitability for chemoproteomic applications, and enantioselective cysteine-labelling. The reactivity of sulfoximine BCBs with electron withdrawing groups on nitrogen is comparable to acrylamides with low to moderate reactivity.
Collapse
Affiliation(s)
- Zhenhao Zhong
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London, W12 0BZ, UK
| | - Brad J W Hocking
- Department of Life Sciences, Imperial College London South Kensington Campus, London, SW7 2AZ, UK
| | - Charles P Brown
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London, W12 0BZ, UK
| | - Tsz-Kan Ma
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London, W12 0BZ, UK
| | - Andrew J P White
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London, W12 0BZ, UK
| | - David J Mann
- Department of Life Sciences, Imperial College London South Kensington Campus, London, SW7 2AZ, UK
| | - Alan Armstrong
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London, W12 0BZ, UK
| | - James A Bull
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London, W12 0BZ, UK
| |
Collapse
|
46
|
Schwalen F, Ghadi C, Ibazizene L, Khan SU, Sopkova-de Oliveira Santos J, Weiswald LB, Voisin-Chiret AS, Meryet-Figuiere M, Kieffer C. UBE2N: Hope on the Cancer Front, How to Inhibit This Promising Target Prospect? J Med Chem 2025; 68:915-928. [PMID: 39806871 DOI: 10.1021/acs.jmedchem.4c01517] [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: 01/16/2025]
Abstract
UBE2N protein belongs to the UE2s family and plays a crucial role in DNA repair, making it an exciting target for the development of innovative anticancer therapies. With the aim of discovering UBE2N inhibitors (UBE2Ni), this perspective seeks to review and provide elements to guide the design of new compounds. We propose a chemoinformatic structural analysis of the protein and its areas of interaction with its different partners. While covalent UBE2Ni are the most advanced molecules in their development, noncovalent inhibitors offer significant advantages that could overcome the limitations of covalent ones, particularly in terms of selectivity. Lastly, to obtain a drug candidate, early assessment of the druggability of compounds is essential in a hit to lead process. For existing UBE2Ni, a critical challenge lies in their pharmacokinetic properties and will obviously have to be considered as early as possible to hope for an application in human therapy.
Collapse
Affiliation(s)
- Florian Schwalen
- Université de Caen Normandie, CERMN UR4258, Normandie Univ, F-14000 Caen, France
- Pharmacie, CHU Caen Normandie, 14033 Caen, France
| | - Côme Ghadi
- Université de Caen Normandie, CERMN UR4258, Normandie Univ, F-14000 Caen, France
| | - Léonie Ibazizene
- Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Université de Caen Normandie, 14076 Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, 14076 Caen, France
| | - Shafi Ullah Khan
- Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Université de Caen Normandie, 14076 Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, 14076 Caen, France
| | | | - Louis-Bastien Weiswald
- Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Université de Caen Normandie, 14076 Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, 14076 Caen, France
| | | | - Matthieu Meryet-Figuiere
- Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Université de Caen Normandie, 14076 Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, 14076 Caen, France
| | - Charline Kieffer
- Université de Caen Normandie, CERMN UR4258, Normandie Univ, F-14000 Caen, France
| |
Collapse
|
47
|
Tamura T, Kawano M, Hamachi I. Targeted Covalent Modification Strategies for Drugging the Undruggable Targets. Chem Rev 2025; 125:1191-1253. [PMID: 39772527 DOI: 10.1021/acs.chemrev.4c00745] [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: 01/11/2025]
Abstract
The term "undruggable" refers to proteins or other biological targets that have been historically challenging to target with conventional drugs or therapeutic strategies because of their structural, functional, or dynamic properties. Drugging such undruggable targets is essential to develop new therapies for diseases where current treatment options are limited or nonexistent. Thus, investigating methods to achieve such drugging is an important challenge in medicinal chemistry. Among the numerous methodologies for drug discovery, covalent modification of therapeutic targets has emerged as a transformative strategy. The covalent attachment of diverse functional molecules to targets provides a powerful platform for creating highly potent drugs and chemical tools as well the ability to provide valuable information on the structures and dynamics of undruggable targets. In this review, we summarize recent examples of chemical methods for the covalent modification of proteins and other biomolecules for the development of new therapeutics and to overcome drug discovery challenges and highlight how such methods contribute toward the drugging of undruggable targets. In particular, we focus on the use of covalent chemistry methods for the development of covalent drugs, target identification, drug screening, artificial modulation of post-translational modifications, cancer specific chemotherapies, and nucleic acid-based therapeutics.
Collapse
Affiliation(s)
- Tomonori Tamura
- Graduate School of Engineering, Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
- Exploratory Research for Advanced Technology (ERATO), Japan Science and Technology Agency (JST), 5 Sanbancho, Chiyoda-ku, Tokyo 102-0075, Japan
| | - Masaharu Kawano
- Graduate School of Engineering, Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Itaru Hamachi
- Graduate School of Engineering, Department of Synthetic Chemistry and Biological Chemistry, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
- Exploratory Research for Advanced Technology (ERATO), Japan Science and Technology Agency (JST), 5 Sanbancho, Chiyoda-ku, Tokyo 102-0075, Japan
| |
Collapse
|
48
|
Takemoto M, Delghandi S, Abo M, Yurimoto K, Odagi M, Singh VP, Wang J, Nakagawa R, Sato SI, Takemoto Y, Farrag AMAS, Kawaguchi Y, Nagasawa K, Honjo T, Chamoto K, Uesugi M. Covalent Plant Natural Product that Potentiates Antitumor Immunity. J Am Chem Soc 2025; 147:2902-2912. [PMID: 39794153 DOI: 10.1021/jacs.4c17837] [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: 01/13/2025]
Abstract
Despite the unprecedented therapeutic potential of immune checkpoint antibody therapies, their efficacy is limited partly by the dysfunction of T cells within the cancer microenvironment. Combination therapies with small molecules have also been explored, but their clinical implementation has been met with significant challenges. To search for antitumor immunity activators, the present study developed a cell-based system that emulates cancer-attenuated T cells. The cell-based screening of 232 natural products containing electrophilic reactive functional groups led to the identification of arvenin I, also known as cucurbitacin B 2-O-β-d-glucoside (CuBg), as a plant natural product that activates T cells within the cancer-competitive environment. Chemoproteomic and mechanistic analyses indicated that arvenin I covalently reacts with and hyperactivates MKK3, thereby reviving the mitochondrial fitness of exhausted T cells through the activation of the p38MAPK pathway. In mice, administration of arvenin I enhanced the efficacy of cancer immunotherapy when used alone or in combination with an immune checkpoint inhibitor. These findings highlight the potential of arvenin I as a covalent kinase activator that potentiates antitumor immunity.
Collapse
Affiliation(s)
- Misao Takemoto
- Division of Biochemistry, Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Sara Delghandi
- Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Masahiro Abo
- Division of Biochemistry, Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Keiko Yurimoto
- Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Minami Odagi
- Department of Biotechnology and Life Science, Graduate School of Technology, Tokyo University of Agriculture and Technology, Koganei, Tokyo 184-8588, Japan
| | - Vaibhav Pal Singh
- Division of Biochemistry, Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Jun Wang
- Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Reiko Nakagawa
- Laboratory for Cell-Free Protein Synthesis, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
| | - Shin-Ichi Sato
- Division of Biochemistry, Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Yasushi Takemoto
- Division of Biochemistry, Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Asmaa M A S Farrag
- Division of Biochemistry, Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Yoshimasa Kawaguchi
- Division of Biochemistry, Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Kazuo Nagasawa
- Department of Biotechnology and Life Science, Graduate School of Technology, Tokyo University of Agriculture and Technology, Koganei, Tokyo 184-8588, Japan
| | - Tasuku Honjo
- Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Kenji Chamoto
- Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
- Department of Immuno-Oncology PDT, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Motonari Uesugi
- Division of Biochemistry, Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8372, Japan
- School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
49
|
Hossain MA, Mayo AK, Ghoshal A, Taft-Benz SA, Anderson EJ, Morales NL, Pressey KD, Vargason AM, Brouwer KLR, Moorman NJ, Heise MT, Willson TM. Species Dependent Metabolism of a Covalent nsP2 Protease Inhibitor with in Vivo Anti-alphaviral Activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632788. [PMID: 39868137 PMCID: PMC11761022 DOI: 10.1101/2025.01.13.632788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
RA-0002034 (1) is a potent covalent inhibitor targeting the alphavirus nsP2 cysteine protease. The species-dependent pharmacokinetics and metabolism of 1 were investigated to evaluate its therapeutic potential. Pharmacokinetic profiling revealed rapid clearance in mice, predominantly mediated by glutathione S-transferase (GST)-catalyzed conjugation. This metabolic liability contrasted with slower clearance observed in human hepatocytes and preclinical species such as rats, dogs, and monkeys. Cross-species studies confirmed the dominance of GST-driven metabolism in mice, whereas oxidative pathways were more pronounced in dogs. Despite rapid systemic clearance, 1 achieved antiviral efficacy in mice, reducing CHIKV viral loads in multiple tissues. Initial estimations of human hepatic clearance and half-life extrapolated from animal data indicate that b.i.d. dosing of 1 will be possible to maintain concentrations sufficient for antiviral activity in humans. These cross-species pharmacokinetic and metabolism studies support the continued evaluation of 1 as a promising anti-alphaviral therapeutic.
Collapse
Affiliation(s)
- Mohammad Anwar Hossain
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Abigail K. Mayo
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Anirban Ghoshal
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sharon A. Taft-Benz
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Elizabeth J. Anderson
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Noah L. Morales
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Katia D. Pressey
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ava M. Vargason
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Eshelman Innovation, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kim L. R. Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nathaniel J. Moorman
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Mark T. Heise
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Timothy M. Willson
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
50
|
Ren J, Zhang Z, Xia Y, Zhao D, Li D, Zhang S. Research Progress on the Structure and Function, Immune Escape Mechanism, Antiviral Drug Development Methods, and Clinical Use of SARS-CoV-2 M pro. Molecules 2025; 30:351. [PMID: 39860219 PMCID: PMC11767629 DOI: 10.3390/molecules30020351] [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: 11/18/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
The three-year COVID-19 pandemic 'has' caused a wide range of medical, social, political, and financial implications. Since the end of 2020, various mutations and variations in SARS-CoV-2 strains, along with the immune escape phenomenon, have emerged. There is an urgent need to identify a relatively stable target for the development of universal vaccines and drugs that can effectively combat both SARS-CoV-2 strains and their mutants. Currently, the main focus in treating SARS-CoV-2 lies in disrupting the virus's life cycle. The main protease (Mpro) is closely associated with virus replication and maturation and plays a crucial role in the early stages of infection. Consequently, it has become an important target for the development of SARS-CoV-2-specific drugs. This review summarizes the recent research progress on the novel coronavirus's main proteases, including the pivotal role of Mpro in the virus's life cycle, the structure and catalytic mechanism of Mpro, the self-maturation mechanism of Mpro, the role of Mpro in virus immune escape, the current methods of developing antiviral drugs targeting Mpro, and the key drugs that have successfully entered clinical trials. The aim is to provide researchers involved in the development of antiviral drugs targeting Mpro with systematic and comprehensive information.
Collapse
Affiliation(s)
| | | | | | | | - Dingqin Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; (J.R.); (Z.Z.); (Y.X.); (D.Z.)
| | - Shujun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; (J.R.); (Z.Z.); (Y.X.); (D.Z.)
| |
Collapse
|