1
|
Hao Y, Zhang B, Chen R. Application of mass spectrometry for the advancement of PROTACs. J Pharm Biomed Anal 2025; 261:116829. [PMID: 40121702 DOI: 10.1016/j.jpba.2025.116829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 01/10/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
The advent of targeted protein degradation technologies, particularly Proteolysis-Targeting Chimeras (PROTACs), enable the selective elimination of target proteins and open up new avenues for the treatment of various diseases. This review delves into the pivotal role of mass spectrometry (MS) in the advancement of PROTACs. MS-based methodologies serve as invaluable tools for identifying PROTAC targets, validating their efficacy, and elucidating ubiquitination sites and protein degradation dynamics. These insights profoundly enrich our comprehension of the mechanisms of action and facilitate the rational design of PROTACs. Furthermore, this review discusses the role of MS in the structural analysis of proteins and the formation of ternary complexes crucial for the activity of PROTACs. The synergy between MS and PROTAC technology holds the promise of groundbreaking advancements in drug discovery by deepening our understanding of the underlying mechanisms that govern PROTAC drug action, thereby promoting the development of innovative strategies for disease treatment.
Collapse
Affiliation(s)
- Yuechen Hao
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Baoshuang Zhang
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Ruibing Chen
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
2
|
Zattoni J, Vottero P, Carena G, Uliveto C, Pozzati G, Morabito B, Gitari E, Tuszynski J, Aminpour M. A comprehensive primer and review of PROTACs and their In Silico design. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 264:108687. [PMID: 40058081 DOI: 10.1016/j.cmpb.2025.108687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/28/2025] [Accepted: 02/25/2025] [Indexed: 04/05/2025]
Abstract
The cutting-edge technique of Proteolysis Targeting Chimeras, or PROTACs, has gained significant attention as a viable approach for specific protein degradation. This innovative technology has vast potential in fields such as cancer therapy and drug development. The development of effective and specific therapies for a range of diseases is within reach with PROTACs, which can target previously "undruggable" proteins while circumventing the off-target effects of conventional small molecule inhibitors. This manuscript aims to discuss the application of in silico techniques to the design of these groundbreaking molecules and develop PROTAC complexes, in order to identify potential PROTAC candidates with favorable drug-like properties. Additionally, this manuscript reviews the strengths and weaknesses of these methods to demonstrate their utility and highlights the challenges and future prospects of in silico PROTAC design. The present review provides a valuable and beginner-friendly resource for researchers and drug developers interested in using in silico methods for PROTAC design, specifically ternary structure prediction.
Collapse
Affiliation(s)
- Jacopo Zattoni
- Department of Biomedical Engineering, University of Alberta, Edmonton, T6G 1Z2, Canada
| | - Paola Vottero
- Department of Biomedical Engineering, University of Alberta, Edmonton, T6G 1Z2, Canada
| | - Gea Carena
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Chiara Uliveto
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Giulia Pozzati
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Benedetta Morabito
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Ebenezea Gitari
- Department of Biochemistry, University of Alberta, Edmonton, T6G 1Z2, Canada
| | - Jack Tuszynski
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; Department of Physics, University of Alberta, 11335 Saskatchewan Dr NW, Edmonton, T6G 2M9, Canada
| | - Maral Aminpour
- Department of Biomedical Engineering, University of Alberta, Edmonton, T6G 1Z2, Canada.
| |
Collapse
|
3
|
Lv W, Jia X, Tang B, Ma C, Fang X, Jin X, Niu Z, Han X. In silico modeling of targeted protein degradation. Eur J Med Chem 2025; 289:117432. [PMID: 40015161 DOI: 10.1016/j.ejmech.2025.117432] [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: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
Targeted protein degradation (TPD) techniques, particularly proteolysis-targeting chimeras (PROTAC) and molecular glue degraders (MGD), have offered novel strategies in drug discovery. With rapid advancement of computer-aided drug design (CADD) and artificial intelligence-driven drug discovery (AIDD) in the biomedical field, a major focus has become how to effectively integrate these technologies into the TPD drug discovery pipeline to accelerate development, shorten timelines, and reduce costs. Currently, the main research directions for applying CADD and AIDD in TPD include: 1) ternary complex modeling; 2) linker generation; 3) strategies to predict degrader targets, activities and ADME/T properties; 4) In silico degrader design and discovery. Models developed in these areas play a crucial role in target identification, drug design, and optimization at various stages of the discovery process. However, the limited size and quality of datasets related to TPD present challenges, leaving room for further improvement in these models. TPD involves the complex ubiquitin-proteasome system, with numerous factors influencing outcomes. Most current models adopt a static perspective to interpret and predict relevant tasks. In the future, it may be necessary to shift toward dynamic approaches that better capture the intricate relationships among these components. Furthermore, incorporating new and diverse chemical spaces will enhance the precision design and application of TPD agents.
Collapse
Affiliation(s)
- Wenxing Lv
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China; Hangzhou Institute of Advanced Technology, Hangzhou, 310000, China.
| | - Xiaojuan Jia
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China.
| | - Bowen Tang
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, China; Guangzhou New Block Technology Co., Ltd., Guangzhou, 510000, China.
| | - Chao Ma
- Guangzhou New Block Technology Co., Ltd., Guangzhou, 510000, China.
| | - Xiaopeng Fang
- Hangzhou Institute of Advanced Technology, Hangzhou, 310000, China.
| | - Xurui Jin
- MindRank AI, Hangzhou, 310000, China.
| | - Zhangming Niu
- MindRank AI, Hangzhou, 310000, China; National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK.
| | - Xin Han
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China; State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Guangxi Normal University), Guilin, 541004, China.
| |
Collapse
|
4
|
Zhu B, Wu Z, Shou Y, Zhao K, Lu Q, Qin JJ, Guo H. Harnessing the Power of Natural Products for Targeted Protein Degradation. Med Res Rev 2025. [PMID: 40304621 DOI: 10.1002/med.22113] [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: 03/27/2025] [Revised: 03/27/2025] [Accepted: 04/02/2025] [Indexed: 05/02/2025]
Abstract
Natural products have garnered significant attention due to their complex chemical structures and remarkable pharmacological activities. With inherent recognition capabilities for protein surfaces, natural products serve as ideal candidates for designing proteolysis-targeting chimeras (PROTACs). The utilization of natural products in PROTAC development offers distinct advantages, including their rich chemical diversity, multitarget activities, and sustainable sourcing. This comprehensive review explores the vast potential of harnessing natural products in PROTAC research. Moreover, the review discusses the application of natural degradant technology, which involves utilizing natural product-based compounds to selectively degrade disease-causing proteins, as well as the implementation of computer-aided drug design (CADD) technology in identifying suitable targets for degradation within the realm of natural products. By harnessing the power of natural products and leveraging computational tools, PROTACs derived from natural products have the potential to revolutionize drug discovery and provide innovative therapeutic interventions for various diseases.
Collapse
Affiliation(s)
- Bo Zhu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
| | - Zheng Wu
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy, Guangxi Medical University, Nanning, Guangxi, China
| | - Yiwen Shou
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy, Guangxi Medical University, Nanning, Guangxi, China
| | - Kaili Zhao
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy, Guangxi Medical University, Nanning, Guangxi, China
| | - Qinpei Lu
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy, Guangxi Medical University, Nanning, Guangxi, China
| | - Jiang-Jiang Qin
- Center for Innovative Drug Research, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Hongwei Guo
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
5
|
Chen J, Wu M, Mo J, Hong J, Wang W, Jin Y, Mao X, Liao X, Li K, Yu X, Chen S, Zeng S, Huang W, Xu H, Wu J, Cao J, Zhou Y, Ying M, Zhu C, He Q, Zhang B, Lin N, Dong X, Che J. Auto-RapTAC: A Versatile and Sustainable Platform for the Automated Rapid Synthesis and Evaluation of PROTAC. J Med Chem 2025; 68:8010-8024. [PMID: 39754574 DOI: 10.1021/acs.jmedchem.4c02438] [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: 01/06/2025]
Abstract
The tedious synthesis and limited throughput biological evaluation remain a great challenge for discovering new proteolysis targeting chimera (PROTAC). To rapidly identify potential PROTAC lead compounds, we report a platform named Auto-RapTAC. Based on the modular characteristic of the PROTAC molecule, a streamlined workflow that integrates lab automation with "click chemistry" joint building-block libraries was constructed. This facilitates the autonomous generation of a variety of PROTACs, each with distinct linkers and E3 ligase ligands, all stored in biocompatible solutions. The ready-for-screening (R4S) approach, when paired with fluorescence-based assays, enables the efficient assessment of the PROTAC degradation activity in a high-throughput manner. To further test the capability of the platform, we identify six new PROTACs that target CDK2, CDK12, and BCL6 within a mere 8-day time frame for each target. In all, this platform could find broad application not only in discovering new PROTACs but also in the rapid development of novel heterobifunctional modalities.
Collapse
Affiliation(s)
- Jiexuan Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mingfei Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jun Mo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ju Hong
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wei Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuheng Jin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinfei Mao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xueyan Liao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Kailin Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaoli Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Sikang Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shenxin Zeng
- Center of Safety Evaluation and Research, School of Pharmacy, Hangzhou Medical College, Hangzhou 310013, China
| | - Wenhai Huang
- Center of Safety Evaluation and Research, School of Pharmacy, Hangzhou Medical College, Hangzhou 310013, China
| | - Hongxia Xu
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jian Wu
- College of Computer Science and Technology, Zhejiang University, Hangzhou 310058, China
| | - Ji Cao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yubo Zhou
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Meidan Ying
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chengliang Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bo Zhang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310024, China
| | - Nengming Lin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310024, China
| | - Xiaowu Dong
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jinxin Che
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
6
|
Salerno A, Wieske LHE, Diehl CJ, Ciulli A. Rational Design of PROTAC Linkers Featuring Ferrocene as a Molecular Hinge to Enable Dynamic Conformational Changes. J Am Chem Soc 2025; 147:13328-13344. [PMID: 40208910 PMCID: PMC12022980 DOI: 10.1021/jacs.4c18354] [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: 12/21/2024] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/12/2025]
Abstract
Proteolysis Targeting Chimeras (PROTACs) are bifunctional molecules that induce ubiquitination and degradation of a target protein via recruitment to an E3 ligase. The linker influences many steps of the PROTAC mode of action, from cellular permeability to ternary complex formation and target degradation. Much interest has therefore been devoted to linker design to fine-tune molecular and mechanistic properties of PROTACs. In this study, we present FerroTACs, a novel PROTAC design strategy incorporating ferrocene as the linker chemotype. We exemplify the approach across three different PROTAC systems: VHL-VHL (homo-PROTACs), VHL-CRBN, and VHL-BETs. We find that ferrocene's unique organometallic structure, featuring freely rotating cyclopentadienyl rings around a central Fe(II) ion, acts as a molecular hinge enabling structural adjustment to the environment that results in properties alteration, i.e., chameleonicity. Conformational analyses via NMR spectroscopy support ferrocene's role in fostering intramolecular interactions that result in a more folded state in an apolar environment. This property promotes compact conformations, improving cellular permeability and reducing efflux liabilities. Cellular assays demonstrate that FerroTACs exhibit robust target degradation and cell permeability profiles, en-par or enhanced compared to benchmark PROTACs CM11, 14a, and MZ1. These findings highlight ferrocene's potential as a new linker design strategy, offering a versatile platform to install and control molecular chameleonicity into next-generation PROTACs.
Collapse
Affiliation(s)
- Alessandra Salerno
- Centre for Targeted Protein
Degradation, School of Life Sciences, University
of Dundee, 1 James Lindsay Place, Dundee DD1 5JJ, U.K.
| | - Lianne H. E. Wieske
- Centre for Targeted Protein
Degradation, School of Life Sciences, University
of Dundee, 1 James Lindsay Place, Dundee DD1 5JJ, U.K.
| | - Claudia J. Diehl
- Centre for Targeted Protein
Degradation, School of Life Sciences, University
of Dundee, 1 James Lindsay Place, Dundee DD1 5JJ, U.K.
| | - Alessio Ciulli
- Centre for Targeted Protein
Degradation, School of Life Sciences, University
of Dundee, 1 James Lindsay Place, Dundee DD1 5JJ, U.K.
| |
Collapse
|
7
|
Guo Q, Yang C, Liu X, Liu J, Zhang W, Lu X, Ding N. Construction of PROTAC molecules by the SuFEx reaction for inducing p300/CBP protein degradation. Bioorg Med Chem 2025; 125:118201. [PMID: 40267748 DOI: 10.1016/j.bmc.2025.118201] [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: 03/04/2025] [Revised: 04/13/2025] [Accepted: 04/15/2025] [Indexed: 04/25/2025]
Abstract
While there have been advancements in the development of innovative PROTACs with sophisticated linkers designed to meet specific requirements, studies on the structure-activity relationships (SAR) of linker length remain a fundamental priority. Although several reliable chemistries for connecting the two ligands-one targeting the protein and the other for E3 ubiquitin ligase-have been established, the potential for utilizing various other methods still needs exploration. In this work, we introduced a concept that employs the SuFEx reaction, a novel family of click chemistry, to quickly construct a small PROTAC library for protein degradation. This was achieved by amidating a sulfonyl fluoride or fluorosulfate precursor (modified with the p300/CBP ligand CPI644) with CRBN ligands that possess amino-carbon chains of varying lengths. The protein degradation effects of the PROTACs created through this strategy were further validated using the p300/CBP overexpressed MDA-MB-468 cell line.
Collapse
Affiliation(s)
- Qiuyu Guo
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Chunxia Yang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Xuyuan Liu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Jian Liu
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wei Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Xiuhong Lu
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| | - Ning Ding
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China.
| |
Collapse
|
8
|
Xie S, Zhu J, Peng Y, Zhan F, Zhan F, He C, Feng D, Xie J, Liu J, Zhu H, Yao H, Xu J, Su Z, Xu S. In Vivo Self-Assembly of PROTACs by Bioorthogonal Chemistry for Precision Cancer Therapy. Angew Chem Int Ed Engl 2025; 64:e202421713. [PMID: 39714400 DOI: 10.1002/anie.202421713] [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: 11/07/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024]
Abstract
Proteolysis targeting chimeras (PROTACs) hold immense promise for targeted protein degradation; however, challenges such as off-target effects, poor drug-likeness properties, and the "hook effect" remain. This study introduces Nano-Click-formed PROTACs (Nano-CLIPTACs) for precise tumor protein degradation in vivo. Traditional PROTACs with high molecular weight were first divided into two smaller druglike precursors capable of self-assembling to form functional PROTACs through a bioorthogonal reaction. Then, optimal CLIPTACs precursors (W4 and Z2) were encapsulated individually into cyclic RGDfC-peptide-modified liposomes to prepare Nano-CLIPTACs, enabling tumor-targeted delivery and subsequent in situ self-assembly to form PROTACs WZ42 within tumor cells. The degradation abilities of Nano-CLIPTACs in vitro and in vivo were further verified using a key oncology target, anaplastic lymphoma kinase (ALK), validating the safety, efficacy and "anti-hook effect" of this strategy. Overall, Nano-CLIPTACs represent a critical step towards the clinical translation of PROTACs technology for precise targeted anti-cancer therapies.
Collapse
Affiliation(s)
- Shaowen Xie
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China
| | - Jingjie Zhu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China
| | - Yihan Peng
- Center of Advanced Pharmaceuticals and Biomaterials, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China
| | - Fangyi Zhan
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China
| | - Feiyan Zhan
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China
| | - Chen He
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China
| | - Dazhi Feng
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China
| | - Jia Xie
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China
| | - Jingyu Liu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China
| | - Huajian Zhu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China
| | - Hong Yao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China
| | - Jinyi Xu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China
| | - Zhigui Su
- Center of Advanced Pharmaceuticals and Biomaterials, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China
| | - Shengtao Xu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China
| |
Collapse
|
9
|
Monsen P, Bommi PV, Grigorescu AA, Lauing KL, Mao Y, Berardi P, Zhai L, Ojo O, Penco-Campillo M, Koch T, Egozi M, Jha S, Dunne SF, Jiang H, Song G, Zhang F, Kregel S, Vaziri-Gohar A, Fanning SW, Sanchez-Gomez P, Allen JM, Yamini B, Lukas RV, Wainwright DA, Schiltz GE. Rational Design and Optimization of a Potent IDO1 Proteolysis Targeting Chimera (PROTAC). J Med Chem 2025; 68:4961-4987. [PMID: 39946350 PMCID: PMC11874035 DOI: 10.1021/acs.jmedchem.5c00026] [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: 01/04/2025] [Revised: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 02/19/2025]
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1) is an immunosuppressive protein that inhibits antitumor immunity through both tryptophan metabolism and nonenzymatic functions. Drugs targeting IDO1 enzyme activity have failed to improve the overall survival of patients with cancer. Developing new therapeutics that neutralize both enzyme- and nonenzyme-derived immunosuppressive IDO1 effects is therefore of high interest. We previously described a novel proteolysis targeting chimera (PROTAC), NU223612, that degrades IDO1 in cultured human glioblastoma (GBM) cells, as well as in well-established brain tumors, in vivo. In this study, we rationally optimized the structure of our lead series to create NU227326, which degrades IDO1 with a DC50 of 5 nM in human GBM cells. Mechanistic studies showed that IDO1 degradation occurred through the ubiquitin-proteasome system and was sustained for at least 2 days, supporting NU227326 as a highly potent IDO1 PROTAC suitable for further studies in GBM and other human cancers.
Collapse
Affiliation(s)
- Paige
J. Monsen
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Prashant V. Bommi
- Department
of Cancer Biology, Loyola University Chicago
Stritch School of Medicine, Maywood, Illinois 60153, United States
| | - Arabela A. Grigorescu
- Department
of Molecular Biosciences, Northwestern University
Weinberg College of Arts and Sciences, Evanston, Illinois 60208, United States
| | - Kristen L. Lauing
- Department
of Cancer Biology, Loyola University Chicago
Stritch School of Medicine, Maywood, Illinois 60153, United States
| | - Yingyu Mao
- High-Throughput
Analysis Laboratory, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, United States
| | - Payton Berardi
- Department
of Cancer Biology, Loyola University Chicago
Stritch School of Medicine, Maywood, Illinois 60153, United States
| | - Lijie Zhai
- Department
of Cancer Biology, Loyola University Chicago
Stritch School of Medicine, Maywood, Illinois 60153, United States
| | - Oluwatomilayo Ojo
- Department
of Cancer Biology, Loyola University Chicago
Stritch School of Medicine, Maywood, Illinois 60153, United States
| | - Manon Penco-Campillo
- Department
of Cancer Biology, Loyola University Chicago
Stritch School of Medicine, Maywood, Illinois 60153, United States
| | - Taylor Koch
- Department
of Cancer Biology, Loyola University Chicago
Stritch School of Medicine, Maywood, Illinois 60153, United States
| | - Michael Egozi
- Department
of Cancer Biology, Loyola University Chicago
Stritch School of Medicine, Maywood, Illinois 60153, United States
| | - Sonam Jha
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Sara F. Dunne
- High-Throughput
Analysis Laboratory, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, United States
| | - Hong Jiang
- HD
Biosciences
(China) Co., Ltd., A WuXi AppTec Company, Shanghai 201201, China
| | - Guiqin Song
- HD
Biosciences
(China) Co., Ltd., A WuXi AppTec Company, Shanghai 201201, China
| | - Fang Zhang
- HD
Biosciences
(China) Co., Ltd., A WuXi AppTec Company, Shanghai 201201, China
| | - Steven Kregel
- Department
of Cancer Biology, Loyola University Chicago
Stritch School of Medicine, Maywood, Illinois 60153, United States
- Cardinal
Bernardin Cancer Center, Maywood, Illinois 60153, United States
| | - Ali Vaziri-Gohar
- Department
of Cancer Biology, Loyola University Chicago
Stritch School of Medicine, Maywood, Illinois 60153, United States
- Cardinal
Bernardin Cancer Center, Maywood, Illinois 60153, United States
- Department
of Surgery, Loyola University Chicago Stritch
School of Medicine, Maywood, Illinois 60153, United States
| | - Sean W. Fanning
- Department
of Cancer Biology, Loyola University Chicago
Stritch School of Medicine, Maywood, Illinois 60153, United States
| | - Pilar Sanchez-Gomez
- Neuro-Oncology
Unit, Unidad Funcional de Investigación
en Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos
III (ISCIII), Madrid 28029, Spain
| | - Jacob M. Allen
- Department
of Health and Kinesiology, University of
Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Bakhtiar Yamini
- Department
of Neurological Surgery, University of Chicago
Medicine, Chicago, Illinois 60637, United States
| | - Rimas V. Lukas
- Department
of Neurology, Northwestern University Feinberg
School of Medicine, Chicago, Illinois 60611, United States
| | - Derek A. Wainwright
- Department
of Cancer Biology, Loyola University Chicago
Stritch School of Medicine, Maywood, Illinois 60153, United States
- Cardinal
Bernardin Cancer Center, Maywood, Illinois 60153, United States
- Department
of Neurological Surgery, Loyola University
Medical Center, Maywood, Illinois 60153, United States
| | - Gary E. Schiltz
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Robert
H. Lurie Comprehensive Cancer Center, Chicago, Illinois 60611, United States
- Department
of Pharmacology, Northwestern University,
Feinberg School of Medicine, Chicago, Illinois 60611 United States
| |
Collapse
|
10
|
McGown A, Vetma V, Crepin D, Lin Y, Adcock C, Craigon C, Nafie J, von Emloh D, Sutton L, Bailey K, Edmunds L, Sharma M, Wilden JD, Coles SJ, Tizzard GJ, Farnaby W, Ciulli A, Kostakis GE, Spencer J. Use of Aldehyde-Alkyne-Amine Couplings to Generate Medicinal Chemistry-Relevant Linkers. ACS Med Chem Lett 2025; 16:278-284. [PMID: 39967623 PMCID: PMC11831382 DOI: 10.1021/acsmedchemlett.4c00531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/20/2025] Open
Abstract
Copper catalyzed aldehyde-alkyne-amine (A3) couplings lead to multifunctional, racemic, propargylic amines, many on a multigram scale. As part of an industrial collaboration, a selection of linkers was purified by chiral HPLC to afford single enantiomers, the absolute configuration of which was determined by vibrational circular dichroism (vCD). To show medicinal chemistry applications, selected linkers were further derivatized into potential cellular probes and (+)-JQ1 containing PROTACs (proteolysis targeting chimeras), which degraded their target protein BRD4.
Collapse
Affiliation(s)
- Andrew McGown
- Sussex
Drug Discovery Centre, School of Life Sciences, University of Sussex, Falmer BN1 9QJ, U.K.
| | - Vesna Vetma
- Centre
for Targeted Protein Degradation, School of Life Sciences, University of Dundee, 1 James Lindsay Place, Dundee DD1 5JJ, U.K.
| | - Damien Crepin
- Sussex
Drug Discovery Centre, School of Life Sciences, University of Sussex, Falmer BN1 9QJ, U.K.
| | - Yan Lin
- Sussex
Drug Discovery Centre, School of Life Sciences, University of Sussex, Falmer BN1 9QJ, U.K.
| | - Claire Adcock
- Sussex
Drug Discovery Centre, School of Life Sciences, University of Sussex, Falmer BN1 9QJ, U.K.
| | - Conner Craigon
- Centre
for Targeted Protein Degradation, School of Life Sciences, University of Dundee, 1 James Lindsay Place, Dundee DD1 5JJ, U.K.
| | - Jordan Nafie
- Biotools,
Inc., 17546 Beeline Highway, Jupiter, Florida 33458, United States
| | - Daniel von Emloh
- Reach
Separations, Biocity,
Pennyfoot Lane, Nottingham NG1 1GF, U.K.
| | - Léa Sutton
- Reach
Separations, Biocity,
Pennyfoot Lane, Nottingham NG1 1GF, U.K.
| | - Kiera Bailey
- Reach
Separations, Biocity,
Pennyfoot Lane, Nottingham NG1 1GF, U.K.
| | - Lewis Edmunds
- Reach
Separations, Biocity,
Pennyfoot Lane, Nottingham NG1 1GF, U.K.
| | - Manvendra Sharma
- Chemistry
Department, School of Life Sciences, University
of Sussex, Falmer BN1 9QJ, U.K.
| | - Jonathan D. Wilden
- Chemistry
Department, School of Life Sciences, University
of Sussex, Falmer BN1 9QJ, U.K.
| | - Simon J. Coles
- National
Crystallography Service Chemistry, University
of Southampton, Southampton SO171BJ, U.K.
| | - Graham J. Tizzard
- National
Crystallography Service Chemistry, University
of Southampton, Southampton SO171BJ, U.K.
| | - William Farnaby
- Centre
for Targeted Protein Degradation, School of Life Sciences, University of Dundee, 1 James Lindsay Place, Dundee DD1 5JJ, U.K.
| | - Alessio Ciulli
- Centre
for Targeted Protein Degradation, School of Life Sciences, University of Dundee, 1 James Lindsay Place, Dundee DD1 5JJ, U.K.
| | - George E. Kostakis
- Chemistry
Department, School of Life Sciences, University
of Sussex, Falmer BN1 9QJ, U.K.
| | - John Spencer
- Sussex
Drug Discovery Centre, School of Life Sciences, University of Sussex, Falmer BN1 9QJ, U.K.
- Chemistry
Department, School of Life Sciences, University
of Sussex, Falmer BN1 9QJ, U.K.
| |
Collapse
|
11
|
Li Y, Zhang X, Xie J, Meng D, Liu M, Chang Y, Feng G, Jiang J, Deng P. Analyzing the Linker Structure of PROTACs throughout the Induction Process: Computational Insights. J Med Chem 2025; 68:3420-3432. [PMID: 39881546 DOI: 10.1021/acs.jmedchem.4c02637] [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: 01/31/2025]
Abstract
Linker structures are a crucial component of proteolysis-targeting chimeras (PROTACs) and have traditionally been designed based on empirical methods, which presents significant challenges in the development of PROTACs. Current optimization strategies typically focus on reducing the number of rotatable bonds in the linker to limit conformational freedom. However, this approach overlooks the complexity of the target protein degradation process. Retrospective analyses suggest that merely adjusting the rotatable bonds in the linker is insufficient to control the conformational freedom of the PROTACs, indicating the need for new optimization strategies. By integration of computational methods such as molecular dynamics simulations, this study investigates the role of the linker throughout the induction process, particularly its impact on the formation and stability of the ternary complex. This approach offers potential for overcoming the limitations of traditional strategies, reducing reliance on empirical methods, and enhancing the overall efficiency and effectiveness of PROTAC design.
Collapse
Affiliation(s)
- Yihao Li
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing 400016, China
- Chongqing Key Research Laboratory for Quality Evaluation and Safety Research of APIs, Chongqing 400016, China
| | - Xiaoxuan Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing 400016, China
- Chongqing Key Research Laboratory for Quality Evaluation and Safety Research of APIs, Chongqing 400016, China
| | - Jiali Xie
- Department of Pharmacy, Mianyang Third People of Hospital, Mianyang 621000 Sichuan, China
| | - Dan Meng
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ming Liu
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Yuxiang Chang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Guangrong Feng
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Junhao Jiang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing 400016, China
- Chongqing Key Research Laboratory for Quality Evaluation and Safety Research of APIs, Chongqing 400016, China
| | - Ping Deng
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing 400016, China
- Chongqing Key Research Laboratory for Quality Evaluation and Safety Research of APIs, Chongqing 400016, China
| |
Collapse
|
12
|
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
|
13
|
Berkley K, Zalejski J, Sharma N, Sharma A. Journey of PROTAC: From Bench to Clinical Trial and Beyond. Biochemistry 2025; 64:563-580. [PMID: 39791901 DOI: 10.1021/acs.biochem.4c00577] [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] [Indexed: 01/12/2025]
Abstract
Proteolysis-targeting chimeras (PROTACs) represent a transformative advancement in drug discovery, offering a method to degrade specific intracellular proteins. Unlike traditional inhibitors, PROTACs are bifunctional molecules that target proteins for elimination, enabling the potential treatment of previously "undruggable" proteins. This concept, pioneered by Crews and his team, introduced the use of small molecules to link a target protein to an E3 ubiquitin ligase, inducing ubiquitination and subsequent degradation of the target protein. By promoting protein degradation rather than merely inhibiting function, PROTACs present a novel therapeutic strategy with enhanced specificity and effectiveness, especially in areas such as cancer and neurodegenerative diseases. Since their initial discovery, the field of PROTAC research has rapidly expanded with numerous PROTACs now designed to target a wide range of disease-relevant proteins. The substantial research, investment, and collaboration across academia and the pharmaceutical industry reflect the growing interest in PROTACs. This Review discusses the journey of PROTACs from initial discovery to clinical trials, highlighting advancements and challenges. Additionally, recent developments in fluorescent and photogenic PROTACs, used for real-time tracking of protein degradation, are presented, showcasing the evolving potential of PROTACs in targeted therapy.
Collapse
Affiliation(s)
- Kyli Berkley
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois 60607, United States
| | - Julian Zalejski
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois 60607, United States
| | - Nidhi Sharma
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois 60607, United States
| | - Ashutosh Sharma
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois 60607, United States
| |
Collapse
|
14
|
Ibrahim S, Umer Khan M, Khurram I, Rehman R, Rauf A, Ahmad Z, Aljohani ASM, Al Abdulmonem W, Quradha MM. Navigating PROTACs in Cancer Therapy: Advancements, Challenges, and Future Horizons. Food Sci Nutr 2025; 13:e70011. [PMID: 39898116 PMCID: PMC11786021 DOI: 10.1002/fsn3.70011] [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: 07/05/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 02/04/2025] Open
Abstract
Proteolysis Targeting Chimeras (PROTACs) have revolutionized cancer therapy by offering a selective and innovative approach to degrade key oncogenic proteins associated with various malignancies. These hybrid molecules exploit the ubiquitin-proteasome system, facilitating the degradation of target proteins through an event-driven mechanism, thereby overcoming drug resistance and enhancing selectivity. With diverse targets including androgen receptors, BTK, estrogen receptors, BET proteins, and BRAF, PROTACs offer a versatile strategy for personalized cancer treatment. Advantages of PROTACs over traditional small molecule inhibitors include their ability to operate at lower concentrations, catalyzing the degradation of multiple proteins of interest with reduced cytotoxicity. Notably, PROTACs address challenges associated with traditionally "undruggable" targets, expanding the therapeutic landscape of cancer therapy. Ongoing preclinical and clinical studies highlight the transformative potential of PROTACs, with promising results in prostate, breast, lung, melanoma, and colorectal cancers. Despite their potential, challenges persist in optimizing physicochemical properties and enhancing bioavailability. Further research is needed to refine PROTAC design and address complexities in molecule development. Nevertheless, the development of oral androgen receptor PROTACs represents a significant milestone, demonstrating the feasibility and efficacy of this innovative therapeutic approach. This review provides a comprehensive overview of PROTACs in cancer therapy, emphasizing their mechanism of action, advantages, and challenges. As PROTAC research progresses, continued exploration in both preclinical and clinical settings will be crucial to unlocking their full therapeutic potential and shaping the future of personalized cancer treatment.
Collapse
Affiliation(s)
- Saooda Ibrahim
- Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
- Centre for Applied Molecular BiologyUniversity of the PunjabLahorePakistan
| | - Muhammad Umer Khan
- Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
| | - Iqra Khurram
- Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
- Centre for Applied Molecular BiologyUniversity of the PunjabLahorePakistan
| | - Raima Rehman
- Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
| | - Abdur Rauf
- Department of ChemistryUniversity of SwabiSwabiKhyber PakhtunkhwaPakistan
| | - Zubair Ahmad
- Department of ChemistryUniversity of SwabiSwabiKhyber PakhtunkhwaPakistan
| | - Abdullah S. M. Aljohani
- Department of Medical BiosciencesCollege of Veterinary Medicine, Qassim UniversityBuraydahSaudi Arabia
| | - Waleed Al Abdulmonem
- Department of PathologyCollege of Medicine, Qassim UniversityBuraydahSaudi Arabia
| | | |
Collapse
|
15
|
London N. Covalent Proximity Inducers. Chem Rev 2025; 125:326-368. [PMID: 39692621 PMCID: PMC11719315 DOI: 10.1021/acs.chemrev.4c00570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024]
Abstract
Molecules that are able to induce proximity between two proteins are finding ever increasing applications in chemical biology and drug discovery. The ability to introduce an electrophile and make such proximity inducers covalent can offer improved properties such as selectivity, potency, duration of action, and reduced molecular size. This concept has been heavily explored in the context of targeted degradation in particular for bivalent molecules, but recently, additional applications are reported in other contexts, as well as for monovalent molecular glues. This is a comprehensive review of reported covalent proximity inducers, aiming to identify common trends and current gaps in their discovery and application.
Collapse
Affiliation(s)
- Nir London
- Department
of Chemical and Structural Biology, The
Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
16
|
Monsen PJ, Bommi PV, Grigorescu AA, Lauing KL, Mao Y, Berardi P, Zhai L, Ojo O, Penco-Campillo M, Koch T, Egozi M, Jha SV, Dunne SF, Jiang H, Song G, Zhang F, Kregel S, Vaziri-Gohar A, Fanning S, Sanchez-Gomez P, Allen JM, Yamini B, Lukas RV, Wainwright DA, Schiltz GE. Rational Design and Optimization of a Potent IDO1 Proteolysis Targeting Chimera (PROTAC). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631731. [PMID: 39829781 PMCID: PMC11741391 DOI: 10.1101/2025.01.07.631731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1) is a potently immunosuppressive protein that inhibits antitumor immunity through both tryptophan metabolism and non-enzymatic functions. Pharmacological therapies targeting IDO1 enzyme activity have generally failed to improve the overall survival of patients with cancer. Developing new therapeutic agents that are capable of neutralizing both enzyme-and non-enzyme-derived immunosuppressive IDO1 effects is therefore of high interest. We previously described the development of a novel Proteolysis Targeting Chimera (PROTAC), NU223612, that degrades IDO1 in cultured human glioblastoma (GBM) cells, as well as in well-established brain tumors, in vivo . In this study, we rationally optimized the composition, rigidity, and linker orientation of the PROTAC structure to create NU227326, which degrades IDO1 with a DC 50 of 5 nM in human GBM cells. Mechanistic studies showed that IDO1 degradation occurred through the ubiquitin-proteasome system and was sustained for at least 2 days, supporting NU227326 as a highly potent IDO1 PROTAC suitable for further studies in GBM and other human cancers.
Collapse
|
17
|
Wang H, Wang Z, Hu L, Yang B, Zong L, Xu D, Yu B, Kong X, Wang M. Discovery of KW0113 as a First and Effective PROTAC Degrader of DNMT1 Protein. ChemMedChem 2024; 19:e202400467. [PMID: 39302251 DOI: 10.1002/cmdc.202400467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
DNA methyltransferase 1 (DNMT1) is an attractive therapeutic target for acute myelocytic leukemia (AML) and other malignancies. It has been reported that the genetic depletion of DNMT1 inhibited AML cell proliferation through reversing DNA methylation abnormalities. However, no DNMT1-targeted PROTAC degraders have been reported yet. Herein, a series of proteolysis-targeting chimera (PROTAC) degrader of DNMT1 based on dicyanopyridine scaffold and VHL E3 ubiquitin ligase ligand was developed. Among them, KW0113 (DC50=643/899 nM in MV4-11/MOLM-13 cells) exhibited optimal DNMT1 degradation. KW0113 induced DNMT1-selective degradation in a dose- and time-dependent manner through VHL engagement. Moreover, KW0113 inhibited AML cell growth by reversing promoter DNA hypermethylation and tumor-suppressor genes silencing. In conclusion, these findings proved the capability of PROTAC strategy for inducing DNMT1 degradation, demonstrated the therapeutic potential of DNMT1-targeted PROTACs. This work also provided a convenient chemical knockdown tool for DNMT1-related studies.
Collapse
Affiliation(s)
- Huihui Wang
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330031, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Zhaoliang Wang
- State Key Laboratory of Respiratory Disease, China-New Zealand Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Linghao Hu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Bingjie Yang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Liangyi Zong
- State Key Laboratory of Respiratory Disease, China-New Zealand Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dounan Xu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Bo Yu
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330031, China
| | - Xiangqian Kong
- State Key Laboratory of Respiratory Disease, China-New Zealand Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mingliang Wang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| |
Collapse
|
18
|
Lin X, Peng C, Liu S, Xiang J, Lin B, Zhan Q, Cao P, Cao T. Modular Assembly of Heterotrifunctional Molecules Enabled by Iodosulfonylation of Allenes and Subsequent Amination. J Med Chem 2024; 67:21251-21263. [PMID: 39565161 DOI: 10.1021/acs.jmedchem.4c02114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Beyond the rapid achievements of therapeutic heterobifunctional molecules, some recent efforts have focused on constructing heterotrifunctional molecules, aiming at developing more potent and selective therapeutic agents or emerging additional functions to heterobifunctional molecules. However, the synthesis of these complex molecules requires a specific design and lengthy steps. We have developed a two-step strategy for the modular construction of heterotrifunctional molecules, enabled by the sustainable and convenient iodosulfonylation of allenes followed by SN2'-selective amination. This strategy successfully incorporates a broad range of biologically active molecules, labeling them with a fluorescent group. The applications of the obtained compounds in selective protein labeling, subcellular imaging, and targeted inhibition of tumor cells make this strategy highly appealing.
Collapse
Affiliation(s)
- Xiaofeng Lin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Chuxiong Peng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Shijie Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Jianpin Xiang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Binyan Lin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Qichen Zhan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Peng Cao
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu, China
- Quzhou People's Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, Zhejiang, China
- Gaoyou Hospital of Traditional Chinese Medicine, Yangzhou 225600, Jiangsu, China
| | - Tao Cao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| |
Collapse
|
19
|
Liu J, Liu Y, Tang J, Gong Q, Yan G, Fan H, Zhang X, Pu C. Recent advances in dual PROTACs degrader strategies for disease treatment. Eur J Med Chem 2024; 279:116901. [PMID: 39341095 DOI: 10.1016/j.ejmech.2024.116901] [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: 08/20/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024]
Abstract
Proteolysis-targeting chimeras (PROTACs) is regarded as an emerging therapeutic strategy with unlimited potential because of its mechanism of inducing target protein degradation though harnessing ubiquitin-proteasome system (UPS). Recently, researchers are combining the advantages of PROTACs and dual-targeted drugs to explore some new types of dual PROTACs degraders. The utilization of dual PROTACs not only enhances the efficiency of selective degradation for two or more distinct proteins, but also facilitates synergistic interactions between target proteins to optimize therapeutic efficacy as well as overcome resistance. In this review, we briefly investigate the innovative strategies of dual degraders based on bivalent or trivalent "Y-type" PROTACs in recent years, outline their design principles, degradation effects, and anticancer activities. Moreover, their advantages and limitations compared with traditional PROTACs will be discussed and provide the outlook on the associated challenges. Meaningfully, the development and application of these dual-targeted PROTACs may point out new directions for replacing numerous combination regimens in the future.
Collapse
Affiliation(s)
- Jianyu Liu
- Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Yanzhuo Liu
- Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Jiao Tang
- Department of Laboratory Medicine, Xindu District People's Hospital, Chengdu, Sichuan, 610500, China
| | - Qianyuan Gong
- Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Guoyi Yan
- School of pharmacy, Xinxiang University, Xinxiang, Henan, 453003, China
| | - Hengrui Fan
- Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Xueping Zhang
- Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, Sichuan, 610041, China.
| | - Chunlan Pu
- Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, 610031, China.
| |
Collapse
|
20
|
Ge J, Hsieh CY, Fang M, Sun H, Hou T. Development of PROTACs using computational approaches. Trends Pharmacol Sci 2024; 45:1162-1174. [PMID: 39567313 DOI: 10.1016/j.tips.2024.10.006] [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: 09/12/2024] [Revised: 10/06/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024]
Abstract
Proteolysis-targeting chimeras (PROTACs) are drugs designed to degrade target proteins via the ubiquitin-proteasome system. With the application of computational biology/chemistry technique in drug design, numerous computer-aided drug design and artificial intelligence (AI)-driven drug design (CADD/AIDD) methods have recently emerged to facilitate the development of PROTAC drugs. We systematically review the role of in silico tools in PROTAC drug design, emphasizing how computational software can model PROTAC action and structure, predict activity, and assist in molecule design. We also discuss current challenges in the rational design of PROTACs from an in silico perspective, such as deviations from small-molecule druggability and the limited availability of training data. We provide an overview of recent discoveries and emerging research in this field, and discuss their potential impact on PROTAC design strategies.
Collapse
Affiliation(s)
- Jingxuan Ge
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China; CarbonSilicon AI Technology Company Ltd, Hangzhou 310018, Zhejiang, China
| | - Chang-Yu Hsieh
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Meijing Fang
- Polytechnic Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Huiyong Sun
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, Jiangsu, China.
| | - Tingjun Hou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China; Polytechnic Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
21
|
Palyada K, Hukkanen R, Leuenroth-Quinn S, Vitsky A, Peterson R, Stamp K, Hoover C, Volak L. Session 5: Protein Degraders. Toxicol Pathol 2024; 52:553-565. [PMID: 39660628 DOI: 10.1177/01926233241300452] [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] [Indexed: 12/12/2024]
Abstract
The so-called undruggable space is an exciting area of potential growth for drug development. Undruggable proteins are defined as those unable to be targeted via conventional small molecule drugs. New modalities are being developed to potentially target these proteins. Targeted protein degradation (TPD) is one such new modality, which over the last two decades has moved from academia to industry. TPD makes use of the endogenous degradation machinery present in all cells, in which E3 ubiquitin ligases mark proteins for degradation via ubiquitin attachment. This session explored the challenges and perspectives of using protein degraders as novel therapeutic agents. The session began with a general introduction to the modality, followed by considerations in evaluating their on- and off-target toxicities including data from an IQ Consortium working group survey. Unique absorption, distribution, metabolism, and excretion (ADME) properties of degrader molecules were presented in relation to their effect on drug development and nonclinical safety assessment. The role of transgenic models in evaluating hemotoxicity associated with cereblon-based therapies was then discussed. A case study to derisk dose-limiting thrombocytopenia was also presented. Finally, a regulatory perspective on the challenges of having toxicity associated with protein degraders was presented.
Collapse
Affiliation(s)
| | | | | | | | | | - Katie Stamp
- Bristol Myers Squibb, Summit, New Jersey, USA
| | | | - Laurie Volak
- Rapport Therapeutics, San Diego, California, USA
| |
Collapse
|
22
|
Cai L, Yue G, Chen Y, Wang L, Yao X, Zou Q, Fu X, Cao D. ET-PROTACs: modeling ternary complex interactions using cross-modal learning and ternary attention for accurate PROTAC-induced degradation prediction. Brief Bioinform 2024; 26:bbae654. [PMID: 39783892 PMCID: PMC11713031 DOI: 10.1093/bib/bbae654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/27/2024] [Accepted: 12/06/2024] [Indexed: 01/12/2025] Open
Abstract
MOTIVATION Accurately predicting the degradation capabilities of proteolysis-targeting chimeras (PROTACs) for given target proteins and E3 ligases is important for PROTAC design. The distinctive ternary structure of PROTACs presents a challenge to traditional drug-target interaction prediction methods, necessitating more innovative approaches. While current state-of-the-art (SOTA) methods using graph neural networks (GNNs) can discern the molecular structure of PROTACs and proteins, thus enabling the efficient prediction of PROTACs' degradation capabilities, they rely heavily on limited crystal structure data of the POI-PROTAC-E3 ternary complex. This reliance underutilizes rich PROTAC experimental data and neglects intricate interaction relationships within ternary complexes. RESULTS In this study, we propose a model based on cross-modal strategy and ternary attention technology, ET-PROTACs, to predict the targeted degradation capabilities of PROTACs. Our model capitalizes on the strengths of cross-modal methods by using equivariant GNN graph neural networks to process the graph structure and spatial coordinates of PROTAC molecules concurrently while utilizing sequence-based methods to learn the protein sequence information. This integration of cross-modal information is cohesively harnessed and channeled into a ternary attention mechanism, specially tailored for the unique structure of PROTACs, enabling the congruent modeling of both PROTAC and protein modalities. Experimental results demonstrate that the ET-PROTACs model outperforms existing SOTA methods. Moreover, visualizing attention scores illuminates crucial residues and atoms pivotal in specific POI-PROTAC-E3 interactions, thus offering invaluable insights and guidance for future pharmaceutical research. AVAILABILITY AND IMPLEMENTATION The codes of our model are available at https://github.com/GuanyuYue/ET-PROTACs.
Collapse
Affiliation(s)
- Lijun Cai
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Guanyu Yue
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Yifan Chen
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Li Wang
- Degree Programs in Systems and Information Engineering, Graduate School of Science and Technology Doctoral Program in Computer Science, University of Tsukuba, Tsukuba, Japan
| | - Xiaojun Yao
- Faculty of Applied Sciences, Centre for Artificial Intelligence Driven Drug Discovery, Macao Polytechnic University, Macao 999078, China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Xiangzheng Fu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, Hunan 410082, China
- Research Institute of Hunan University in Chongqing, Chongqing 401120, China
| | - Dongsheng Cao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410003, China
| |
Collapse
|
23
|
Mi D, Li C, Li Y, Yao M, Li Y, Hong K, Xie C, Chen Y. Discovery of novel BCL6-Targeting PROTACs with effective antitumor activities against DLBCL in vitro and in vivo. Eur J Med Chem 2024; 277:116789. [PMID: 39208743 DOI: 10.1016/j.ejmech.2024.116789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/09/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
The transcriptional repressor B cell lymphoma 6 (BCL6) plays a critical role in driving tumorigenesis of diffuse large B-cell lymphoma (DLBCL). However, the therapeutic potential of inhibiting or degrading BCL6 for DLBCL has not been thoroughly understood. Herein, we reported the discovery of a series of novel BCL6-targeting PROTACs based on our previously reported N-phenyl-4-pyrimidinamine BCL6 inhibitors. The optimal compound DZ-837 degraded BCL6 with DC50 values around 600 nM and effectively inhibited the proliferation of several DLBCL cell lines. Further study indicated that DZ-837 induced significant G1 phase arrest and exhibited sustained reactivation of BCL6 downstream genes. In the SU-DHL-4 xenograft model, DZ-837 significantly inhibited tumor growth with TGI of 71.8 % at 40 mg/kg once daily. Furthermore, the combination of DZ-837 with BTK inhibitor Ibrutinib showed synergistic effects and overcame acquired resistance against DLBCL cells. Overall, our findings demonstrate that DZ-837 is an effective BCL6 degrader for DLBCL treatment as a monotherapy or in combination with Ibrutinib.
Collapse
MESH Headings
- Humans
- Proto-Oncogene Proteins c-bcl-6/antagonists & inhibitors
- Proto-Oncogene Proteins c-bcl-6/metabolism
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/chemical synthesis
- Animals
- Cell Proliferation/drug effects
- Mice
- Structure-Activity Relationship
- Drug Discovery
- Drug Screening Assays, Antitumor
- Molecular Structure
- Dose-Response Relationship, Drug
- Cell Line, Tumor
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/metabolism
- Proteolysis Targeting Chimera
Collapse
Affiliation(s)
- Dazhao Mi
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Cheng Li
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yuzhan Li
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | | | - Yan Li
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China
| | - Keyu Hong
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China; Lingang Laboratory, Shanghai, 200031, China
| | - Chengying Xie
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China; Lingang Laboratory, Shanghai, 200031, China.
| | - Yihua Chen
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China; School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunna, 650500, China; Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, Yunnan, 650500, China.
| |
Collapse
|
24
|
Ning Y, Zhu Z, Wang Y, Fan X, Wang J, Qian H, Qiu X, Wang Y. Design, synthesis, and biological evaluation of RSL3-based GPX4 degraders with hydrophobic tags. Eur J Med Chem 2024; 277:116719. [PMID: 39094276 DOI: 10.1016/j.ejmech.2024.116719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/17/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
Ferroptosis is a new type of programmed cell death characterized by iron-dependent lipid peroxidation, during which glutathione peroxidase 4 (GPX4) plays an essential role and is well-recognized as a promising therapeutic target for cancer treatment. Although some GPX4 degradation molecules have been developed to induce ferroptosis, the discovery of GPX4 degraders with hydrophobic tagging (HyT) as an innovative approach is more challenging. Herein, we designed and synthesized a series of HyT degraders by linking the GPX4 inhibitor RSL3 with a hydrophobic and bulky group of adamantane. Among them, compound R8 is a potent degrader (DC50, 24h = 0.019 μM) which can effectively degrade GPX4 in a dose- and time-dependent manner. Furthermore, compound R8 exhibited superior in vitro antitumor potency against HT1080 and MDA-MB-231 cell lines with IC50 values of 24 nM and 32 nM respectively, which are 4 times more potent than parental compound RSL3. Mechanistic investigation evidenced that R8 consumes GPX4 protein mainly through the ubiquitin proteasome (UPS) and enables to induce the accumulation of LPO, thereby triggering ferroptosis. Our work presented the novel GPX4 degrader of R8 by HyT strategy, and provided a promising pathway of degradation agents for the treatment of ferroptosis relevant diseases.
Collapse
Affiliation(s)
- Yao Ning
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, Shandong, PR China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, PR China
| | - Zeqi Zhu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, Shandong, PR China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, PR China
| | - Yicheng Wang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, Shandong, PR China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, PR China
| | - Xuejing Fan
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, Shandong, PR China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, PR China
| | - Jing Wang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, Shandong, PR China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, PR China
| | - Huimei Qian
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, Shandong, PR China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, PR China
| | - Xue Qiu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, Shandong, PR China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, PR China
| | - Yong Wang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, Shandong, PR China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, PR China.
| |
Collapse
|
25
|
Zhang SH, Zeng N, Xu JZ, Liu CQ, Xu MY, Sun JX, An Y, Zhong XY, Miao LT, Wang SG, Xia QD. Recent breakthroughs in innovative elements, multidimensional enhancements, derived technologies, and novel applications of PROTACs. Biomed Pharmacother 2024; 180:117584. [PMID: 39427546 DOI: 10.1016/j.biopha.2024.117584] [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: 08/06/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024] Open
Abstract
Proteolysis Targeting Chimera (PROTAC) is an emerging and evolving technology based on targeted protein degradation (TPD). Small molecule PROTACs have shown great efficacy in degrading disease-specific proteins in preclinical and clinical studies, but also showed various limitations. In recent years, new technologies and advances in TPD have provided additional optimized strategies based on conventional PROTACs that can overcome the shortcomings of conventional PROTACs in terms of undruggable targets, bioavailability, tissue-specificity, spatiotemporal control, and degradation scope. In addition, some designs of special targeting chimeras and applications based on multidisciplinary science have shed light on novel therapeutic modalities and drug design. However, each improvement has its own advantages, disadvantages and application conditions. In this review, we summarize the exploration of PROTAC elements, depict a landscape of improvements and derived concepts of PROTACs, and expect to provide perspectives for technological innovations, combinations and applications in future targeting chimera design.
Collapse
Affiliation(s)
- Si-Han Zhang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Na Zeng
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Jin-Zhou Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Chen-Qian Liu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Meng-Yao Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Jian-Xuan Sun
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Ye An
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Xing-Yu Zhong
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Lin-Tao Miao
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Shao-Gang Wang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China.
| | - Qi-Dong Xia
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China.
| |
Collapse
|
26
|
Qiu Y, Wiewiora RP, Izaguirre JA, Xu H, Sherman W, Tang W, Huang X. Non-Markovian Dynamic Models Identify Non-Canonical KRAS-VHL Encounter Complex Conformations for Novel PROTAC Design. JACS AU 2024; 4:3857-3868. [PMID: 39483225 PMCID: PMC11522902 DOI: 10.1021/jacsau.4c00503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/26/2024] [Accepted: 09/16/2024] [Indexed: 11/03/2024]
Abstract
Targeted protein degradation (TPD) is emerging as a promising therapeutic approach for cancer and other diseases, with an increasing number of programs demonstrating its efficacy in human clinical trials. One notable method for TPD is Proteolysis Targeting Chimeras (PROTACs) that selectively degrade a protein of interest (POI) through E3-ligase induced ubiquitination followed by proteasomal degradation. PROTACs utilize a warhead-linker-ligand architecture to bring the POI (bound to the warhead) and the E3 ligase (bound to the ligand) into proximity. The resulting non-native protein-protein interactions (PPIs) formed between the POI and E3 ligase lead to the formation of a stable ternary complex, enhancing cooperativity for TPD. A significant challenge in PROTAC design is the screening of the linkers to induce favorable non-native PPIs between POI and E3 ligase. Here, we present a physics-based computational protocol to predict noncanonical and metastable PPI interfaces between an E3 ligase and a given POI, aiding in the design of linkers to stabilize the ternary complex and enhance degradation. Specifically, we build the non-Markovian dynamic model using the Integrative Generalized Master equation (IGME) method from ∼1.5 ms all-atom molecular dynamics simulations of linker-less encounter complex, to systematically explore the inherent PPIs between the oncogene homologue protein and the von Hippel-Lindau E3 ligase. Our protocol revealed six metastable states each containing a different PPI interface. We selected three of these metastable states containing promising PPIs for linker design. Our selection criterion included thermodynamic and kinetic stabilities of PPIs and the accessibility between the solvent-exposed sites on the warheads and E3 ligand. One selected PPIs closely matches a recent cocrystal PPI interface structure induced by an experimentally designed PROTAC with potent degradation efficacy. We anticipate that our protocol has significant potential for widespread application in predicting metastable POI-ligase interfaces that can enable rational design of PROTACs.
Collapse
Affiliation(s)
- Yunrui Qiu
- Department
of Chemistry, Theoretical Chemistry Institute, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
- Data
Science Institute, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | | | | | - Huafeng Xu
- Atommap
Corporation, NY, New York 10013, United
States
| | - Woody Sherman
- Psivant
Therapeutics, Boston, Massachusetts 02210, United States
| | - Weiping Tang
- Lachman
Institute for Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Xuhui Huang
- Department
of Chemistry, Theoretical Chemistry Institute, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
- Data
Science Institute, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
27
|
Yan S, Zhang G, Luo W, Xu M, Peng R, Du Z, Liu Y, Bai Z, Xiao X, Qin S. PROTAC technology: From drug development to probe technology for target deconvolution. Eur J Med Chem 2024; 276:116725. [PMID: 39083982 DOI: 10.1016/j.ejmech.2024.116725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/19/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
Drug development remains a critical focus within the global pharmaceutical industry. To date, more than 80 % of disease targets are considered difficult to target. The emergence of PROTAC technology has, to some extent, alleviated this challenge. Since introduction, PROTAC technology has evolved through the peptide E3 ligase ligand phase and the small molecule E3 ligase ligand phase. Currently, multiple PROTAC molecules are in the clinical research phase, showing promising potential for addressing drug resistance, disease recurrence, and intractable targets. Target deconvolution is a crucial step in the drug discovery and development process. Due to the exceptional targeting ability and specificity of PROTAC, it is widely used and promoted as an innovative technology for discovering new drug targets, leading to significant breakthroughs. The use of PROTAC probe requires only a catalytic dose and weak interaction with the target protein to achieve target degradation. Thus, it offers substantial advantages over traditional probes, particularly in identifying new targets that are low-abundance or difficult to target. This review provides a comprehensive overview of the advancements made by PROTAC technology in drug development and drug target discovery, while also systematically reviewing the workflow of PROTAC probe. With the ongoing development of PROTAC technology, PROTAC probe is poised to become a key research area in future drug target deconvolution.
Collapse
Affiliation(s)
- Si Yan
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Research Center for Precision Medication of Chinese Medicine, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha, PR China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, PR China; Department of Hepatology, China Military Institute of Chinese Materia, The Fifth Medical Center of PLA General Hospital, Beijing, PR China
| | - Guangshuai Zhang
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Research Center for Precision Medication of Chinese Medicine, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha, PR China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, PR China; Department of Hepatology, China Military Institute of Chinese Materia, The Fifth Medical Center of PLA General Hospital, Beijing, PR China
| | - Wei Luo
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Research Center for Precision Medication of Chinese Medicine, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha, PR China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, PR China
| | - Mengwei Xu
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Research Center for Precision Medication of Chinese Medicine, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha, PR China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, PR China
| | - Rui Peng
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Research Center for Precision Medication of Chinese Medicine, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha, PR China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, PR China
| | - Ziwei Du
- Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, PR China
| | - Yan Liu
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Research Center for Precision Medication of Chinese Medicine, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha, PR China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, PR China
| | - Zhaofang Bai
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Research Center for Precision Medication of Chinese Medicine, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha, PR China; Department of Hepatology, China Military Institute of Chinese Materia, The Fifth Medical Center of PLA General Hospital, Beijing, PR China.
| | - Xiaohe Xiao
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Research Center for Precision Medication of Chinese Medicine, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha, PR China; Department of Hepatology, China Military Institute of Chinese Materia, The Fifth Medical Center of PLA General Hospital, Beijing, PR China.
| | - Shuanglin Qin
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Research Center for Precision Medication of Chinese Medicine, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha, PR China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, PR China; Department of Hepatology, China Military Institute of Chinese Materia, The Fifth Medical Center of PLA General Hospital, Beijing, PR China.
| |
Collapse
|
28
|
Bálint D, Póti ÁL, Alexa A, Sok P, Albert K, Torda L, Földesi-Nagy D, Csókás D, Turczel G, Imre T, Szarka E, Fekete F, Bento I, Bojtár M, Palkó R, Szabó P, Monostory K, Pápai I, Soós T, Reményi A. Reversible covalent c-Jun N-terminal kinase inhibitors targeting a specific cysteine by precision-guided Michael-acceptor warheads. Nat Commun 2024; 15:8606. [PMID: 39366946 PMCID: PMC11452492 DOI: 10.1038/s41467-024-52573-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/13/2024] [Indexed: 10/06/2024] Open
Abstract
There has been a surge of interest in covalent inhibitors for protein kinases in recent years. Despite success in oncology, the off-target reactivity of these molecules is still hampering the use of covalent warhead-based strategies. Herein, we disclose the development of precision-guided warheads to mitigate the off-target challenge. These reversible warheads have a complex and cyclic structure with optional chirality center and tailored steric and electronic properties. To validate our proof-of-concept, we modified acrylamide-based covalent inhibitors of c-Jun N-terminal kinases (JNKs). We show that the cyclic warheads have high resilience against off-target thiols. Additionally, the binding affinity, residence time, and even JNK isoform specificity can be fine-tuned by adjusting the substitution pattern or using divergent and orthogonal synthetic elaboration of the warhead. Taken together, the cyclic warheads presented in this study will be a useful tool for medicinal chemists for the deliberate design of safer and functionally fine-tuned covalent inhibitors.
Collapse
Affiliation(s)
- Dániel Bálint
- Organocatalysis Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
- Hevesy György PhD School of Chemistry, Eötvös Loránd University, 1117, Budapest, Hungary
| | - Ádám Levente Póti
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
- Doctoral School of Biology, Eötvös Loránd University, 1117, Budapest, Hungary
| | - Anita Alexa
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Péter Sok
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Krisztián Albert
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Lili Torda
- Organocatalysis Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Dóra Földesi-Nagy
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Dániel Csókás
- Theoretical Chemistry Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Gábor Turczel
- NMR Research Laboratory, Centre for Structural Science, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Tímea Imre
- MS Metabolomic Research Laboratory, Centre for Structural Science, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Eszter Szarka
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Ferenc Fekete
- Metabolic Drug-interactions Research Group, Institute of Molecular Life Sciences, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Isabel Bento
- European Molecular Biology Laboratory, EMBL, Hamburg, Germany
| | - Márton Bojtár
- Chemical Biology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Roberta Palkó
- Organocatalysis Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Pál Szabó
- MS Metabolomic Research Laboratory, Centre for Structural Science, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Katalin Monostory
- Metabolic Drug-interactions Research Group, Institute of Molecular Life Sciences, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Imre Pápai
- Theoretical Chemistry Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Tibor Soós
- Organocatalysis Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary.
| | - Attila Reményi
- Biomolecular Interaction Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117, Budapest, Hungary.
| |
Collapse
|
29
|
Su Y, Ngea GLN, Wang K, Lu Y, Godana EA, Ackah M, Yang Q, Zhang H. Deciphering the mechanism of E3 ubiquitin ligases in plant responses to abiotic and biotic stresses and perspectives on PROTACs for crop resistance. PLANT BIOTECHNOLOGY JOURNAL 2024; 22:2811-2843. [PMID: 38864414 PMCID: PMC11536463 DOI: 10.1111/pbi.14407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/12/2024] [Accepted: 05/27/2024] [Indexed: 06/13/2024]
Abstract
With global climate change, it is essential to find strategies to make crops more resistant to different stresses and guarantee food security worldwide. E3 ubiquitin ligases are critical regulatory elements that are gaining importance due to their role in selecting proteins for degradation in the ubiquitin-proteasome proteolysis pathway. The role of E3 Ub ligases has been demonstrated in numerous cellular processes in plants responding to biotic and abiotic stresses. E3 Ub ligases are considered a class of proteins that are difficult to control by conventional inhibitors, as they lack a standard active site with pocket, and their biological activity is mainly due to protein-protein interactions with transient conformational changes. Proteolysis-targeted chimeras (PROTACs) are a new class of heterobifunctional molecules that have emerged in recent years as relevant alternatives for incurable human diseases like cancer because they can target recalcitrant proteins for destruction. PROTACs interact with the ubiquitin-proteasome system, principally the E3 Ub ligase in the cell, and facilitate proteasome turnover of the proteins of interest. PROTAC strategies harness the essential functions of E3 Ub ligases for proteasomal degradation of proteins involved in dysfunction. This review examines critical advances in E3 Ub ligase research in plant responses to biotic and abiotic stresses. It highlights how PROTACs can be applied to target proteins involved in plant stress response to mitigate pathogenic agents and environmental adversities.
Collapse
Affiliation(s)
- Yingying Su
- School of Food and Biological EngineeringJiangsu UniversityZhenjiangChina
| | - Guillaume Legrand Ngolong Ngea
- School of Food and Biological EngineeringJiangsu UniversityZhenjiangChina
- Institute of Fisheries Sciences, University of DoualaDoualaCameroon
| | - Kaili Wang
- School of Food and Biological EngineeringJiangsu UniversityZhenjiangChina
| | - Yuchun Lu
- School of Food and Biological EngineeringJiangsu UniversityZhenjiangChina
| | - Esa Abiso Godana
- School of Food and Biological EngineeringJiangsu UniversityZhenjiangChina
| | - Michael Ackah
- School of Food and Biological EngineeringJiangsu UniversityZhenjiangChina
| | - Qiya Yang
- School of Food and Biological EngineeringJiangsu UniversityZhenjiangChina
| | - Hongyin Zhang
- School of Food and Biological EngineeringJiangsu UniversityZhenjiangChina
| |
Collapse
|
30
|
Dong Y, Ma T, Xu T, Feng Z, Li Y, Song L, Yao X, Ashby CR, Hao GF. Characteristic roadmap of linker governs the rational design of PROTACs. Acta Pharm Sin B 2024; 14:4266-4295. [PMID: 39525578 PMCID: PMC11544172 DOI: 10.1016/j.apsb.2024.04.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 02/11/2024] [Accepted: 04/02/2024] [Indexed: 11/16/2024] Open
Abstract
Proteolysis targeting chimera (PROTAC) technology represents a groundbreaking development in drug discovery, leveraging the ubiquitin‒proteasome system to specifically degrade proteins responsible for the disease. PROTAC is characterized by its unique heterobifunctional structure, which comprises two functional domains connected by a linker. The linker plays a pivotal role in determining PROTAC's biodegradative efficacy. Advanced and rationally designed functional linkers for PROTAC are under development. Nonetheless, the correlation between linker characteristics and PROTAC efficacy remains under-investigated. Consequently, this study will present a multidisciplinary analysis of PROTAC linkers and their impact on efficacy, thereby guiding the rational design of linkers. We will primarily discuss the structural types and characteristics of PROTAC linkers, and the optimization strategies used for their rational design. Furthermore, we will discuss how factors like linker length, group type, flexibility, and linkage site affect the biodegradation efficiency of PROTACs. We believe that this work will contribute towards the advancement of rational linker design in the PROTAC research area.
Collapse
Affiliation(s)
- Yawen Dong
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Tingting Ma
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Ting Xu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Zhangyan Feng
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Yonggui Li
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Lingling Song
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Xiaojun Yao
- Faculty of Applied Sciences, Macau Polytechnic University, Macau 999078, China
| | - Charles R. Ashby
- Department of Pharmaceutical Sciences, St. John's University, New York, NY 11439, USA
| | - Ge-Fei Hao
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals, Guizhou University, Guiyang 550025, China
| |
Collapse
|
31
|
Chen Y, Liu F, Pal S, Hu Q. Proteolysis-targeting drug delivery system (ProDDS): integrating targeted protein degradation concepts into formulation design. Chem Soc Rev 2024; 53:9582-9608. [PMID: 39171633 DOI: 10.1039/d4cs00411f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Targeted protein degradation (TPD) has emerged as a revolutionary paradigm in drug discovery and development, offering a promising avenue to tackle challenging therapeutic targets. Unlike traditional drug discovery approaches that focus on inhibiting protein function, TPD aims to eliminate proteins of interest (POIs) using modular chimeric structures. This is achieved through the utilization of proteolysis-targeting chimeras (PROTACs), which redirect POIs to E3 ubiquitin ligases, rendering them for degradation by the cellular ubiquitin-proteasome system (UPS). Additionally, other TPD technologies such as lysosome-targeting chimeras (LYTACs) and autophagy-based protein degraders facilitate the transportation of proteins to endo-lysosomal or autophagy-lysosomal pathways for degradation, respectively. Despite significant growth in preclinical TPD research, many chimeras fail to progress beyond this stage in the drug development. Various factors contribute to the limited success of TPD agents, including a significant hurdle of inadequate delivery to the target site. Integrating TPD into delivery platforms could surmount the challenges of in vivo applications of TPD strategies by reshaping their pharmacokinetics and pharmacodynamic profiles. These proteolysis-targeting drug delivery systems (ProDDSs) exhibit superior delivery performance, enhanced targetability, and reduced off-tissue side effects. In this review, we will survey the latest progress in TPD-inspired drug delivery systems, highlight the importance of introducing delivery ideas or technologies to the development of protein degraders, outline design principles of protein degrader-inspired delivery systems, discuss the current challenges, and provide an outlook on future opportunities in this field.
Collapse
Affiliation(s)
- Yu Chen
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Fengyuan Liu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Samira Pal
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
32
|
Pierri M, Liu X, Kroupova A, Rutter Z, Hallatt AJ, Ciulli A. Stereochemical inversion at a 1,4-cyclohexyl PROTAC linker fine-tunes conformation and binding affinity. Bioorg Med Chem Lett 2024; 110:129861. [PMID: 38942127 DOI: 10.1016/j.bmcl.2024.129861] [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: 03/24/2024] [Revised: 06/16/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
Proteolysis targeting chimeras (PROTACs) are heterobifunctional small-molecule degraders made of a linker connecting a target-binding moiety to a ubiquitin E3 ligase-binding moiety. The linker unit is known to influence the physicochemical and pharmacokinetic properties of PROTACs, as well as the properties of ternary complexes, in turn impacting on their degradation activity in cells and in vivo. Our LRRK2 PROTAC XL01126, bearing a trans-cyclohexyl group in the linker, is a better and more cooperative degrader than its corresponding cis- analogue despite its much weaker binary binding affinities. Here, we investigate how this subtle stereocenter alteration in the linker affects the ligand binding affinity to the E3 ligase VHL. We designed a series of molecular matched pairs, truncating from the full PROTACs down to the VHL ligand, and find that across the series the trans-cyclohexyl compounds showed consistently weaker VHL-binding affinity compared to the cis- counterparts. High-resolution co-crystal structures revealed that the trans linker exhibits a rigid stick-out conformation, while the cis linker collapses into a folded-back conformation featuring a network of intramolecular contacts and long-range interactions with VHL. These observations are noteworthy as they reveal how a single stereochemical inversion within a PROTAC linker impacts conformational rigidity and binding mode, in turn fine-tuning differentiated propensity to binary and ternary complex formation, and ultimately cellular degradation activity.
Collapse
Affiliation(s)
- Martina Pierri
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, 1 James Lindsay Place, DD1 5JJ Dundee, United Kingdom
| | - Xingui Liu
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, 1 James Lindsay Place, DD1 5JJ Dundee, United Kingdom
| | - Alena Kroupova
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, 1 James Lindsay Place, DD1 5JJ Dundee, United Kingdom
| | - Zoe Rutter
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, 1 James Lindsay Place, DD1 5JJ Dundee, United Kingdom
| | - Alex J Hallatt
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, 1 James Lindsay Place, DD1 5JJ Dundee, United Kingdom
| | - Alessio Ciulli
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, 1 James Lindsay Place, DD1 5JJ Dundee, United Kingdom.
| |
Collapse
|
33
|
Zhang J, Chen X, Chen C, Li F, Song X, Liu C, Liao K, Su MY, Tan CSH, Fang L, Rao H. Distinct Amino Acid-Based PROTACs Target Oncogenic Kinases for Degradation in Non-Small Cell Lung Cancer (NSCLC). J Med Chem 2024; 67:13666-13680. [PMID: 39114932 DOI: 10.1021/acs.jmedchem.4c00208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Proteolysis-targeting chimeras (PROTACs) selectively eliminate detrimental proteins by exploiting the ubiquitin-proteasome system (UPS), representing a promising therapeutic strategy against various diseases. Effective adaptations of degradation signal sequences and E3 ligases for PROTACs remain limited. Here, we employed three amino acids─Gly, Pro, and Lys─as the ligand to recruit the corresponding E3 ligases: CRL2ZYG11B/ZER1, GID4, and UBRs, to degrade EML4-ALK and mutant EGFR, two oncogenic drivers in NSCLC. We found that the extent of EML4-ALK and EGFR reduction can be easily fine-tuned by using different degradation signals. These amino acid-based PROTACs, termed AATacs, hindered proliferation and induced cell cycle arrest and apoptosis of NSCLC cells in vitro. Compared to other PROTACs, AATacs are small, interchangeable but with different degradation efficiency. Our study further expands the repertoire of E3 ligases and their ligands for PROTAC application, improving the versatility and utility of targeted protein degradation for therapeutic purposes.
Collapse
Affiliation(s)
- Jianchao Zhang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xiao Chen
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Congli Chen
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China
| | - Fengming Li
- Department of Chemistry, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xiaoxiao Song
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Chaowei Liu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Kefan Liao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ming-Yuan Su
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen 518055, China
| | - Chris Soon Heng Tan
- Department of Chemistry, Southern University of Science and Technology, Shenzhen 518055, China
| | - Lijing Fang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China
| | - Hai Rao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
34
|
Wu M, Wu Y, Jin Y, Mao X, Zeng S, Yu H, Zhang J, Jin Y, Wu Y, Xu T, Chen Y, Wang Y, Yao X, Che J, Huang W, Dong X. Discovery of an Exceptionally Orally Bioavailable and Potent HPK1 PROTAC with Enhancement of Antitumor Efficacy of Anti-PD-L1 Therapy. J Med Chem 2024; 67:13852-13878. [PMID: 39084610 DOI: 10.1021/acs.jmedchem.4c00644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
HPK1, a well-known negative regulator of T cell receptors, can cause T cell dysfunction when abnormally activated. In this study, a PROTAC C3 was designed and synthesized by optimizing the physicochemical properties of the warhead, linker, and CRBN ligand. C3 demonstrated significant HPK1 degradation with a DC50 of 21.26 nM, excellent oral absorption with a Cmax of 10,899.92 ng/mL, and a bioavailability (F %) of 81.7%. C3 also showed degradation selectivity and potent immune activation effects. Proteomic and WB analyses revealed that immune-activating effect of C3 is attributed to the inhibition of SLP76 and NF-κB signaling pathways, as well as the enhancement of MAPK signaling pathway transduction. In vivo efficacy study demonstrated that oral administration of C3 in combination with anti-PDL1 antibody significantly inhibited tumor growth (tumor growth inhibition = 65.58%). These findings suggest that C3, a novel HPK1 PROTAC, holds promise as a therapeutic agent for tumor immunotherapy.
Collapse
Affiliation(s)
- Mingfei Wu
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Yiquan Wu
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Yuyuan Jin
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou 310058, P. R. China
| | - Xinfei Mao
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Shenxin Zeng
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou 310058, P. R. China
| | - Hengyuan Yu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Jingyu Zhang
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Yuheng Jin
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Yizhe Wu
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Tengfei Xu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Yong Chen
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Yuwei Wang
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
| | - Xiaojun Yao
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macau 999078, P. R. China
| | - Jinxin Che
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Wenhai Huang
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou 310058, P. R. China
| | - Xiaowu Dong
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou 310058, P. R. China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, P. R. China
| |
Collapse
|
35
|
Feng Y, Hu X, Wang X. Targeted protein degradation in hematologic malignancies: clinical progression towards novel therapeutics. Biomark Res 2024; 12:85. [PMID: 39169396 PMCID: PMC11340087 DOI: 10.1186/s40364-024-00638-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024] Open
Abstract
Targeted therapies, such as small molecule kinase inhibitors, have made significant progress in the treatment of hematologic malignancies by directly modulating protein activity. However, issues such as drug toxicity, drug resistance due to target mutations, and the absence of key active sites limit the therapeutic efficacy of these drugs. Targeted protein degradation (TPD) presents an emergent and rapidly evolving therapeutic approach that selectively targets proteins of interest (POI) based on endogenous degradation processes. With an event-driven pharmacology of action, TPD achieves efficacy with catalytic amounts, avoiding drug-related toxicity. Furthermore, TPD has the unique mode of degrading the entire POI, such that resistance derived from mutations in the targeted protein has less impact on its degradation function. Proteolysis-targeting chimeras (PROTACs) and molecular glue degraders (MGDs) are the most maturely developed TPD techniques. In this review, we focus on both preclinical experiments and clinical trials to provide a comprehensive summary of the safety and clinical effectiveness of PROTACs and MGDs in hematologic malignancies over the past two decades. In addition, we also delineate the challenges and opportunities associated with these burgeoning degradation techniques. TPD, as an approach to the precise degradation of specific proteins, provides an important impetus for its future application in the treatment of patients with hematologic malignancies.
Collapse
Affiliation(s)
- Yupiao Feng
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Xinting Hu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
- Taishan Scholars Program of Shandong Province, Jinan, Shandong, 250021, China.
| |
Collapse
|
36
|
Martin PL, Pérez-Areales FJ, Rao SV, Walsh SJ, Carroll JS, Spring DR. Towards the Targeted Protein Degradation of PRMT1. ChemMedChem 2024; 19:e202400269. [PMID: 38724444 DOI: 10.1002/cmdc.202400269] [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: 04/15/2024] [Revised: 05/09/2024] [Indexed: 07/21/2024]
Abstract
Targeting the protein arginine methyltransferase 1 (PRMT1) has emerged as a promising therapeutic strategy in cancer treatment. The phase 1 clinical trial for GSK3368715, the first PRMT1 inhibitor to enter the clinic, was terminated early due to a lack of clinical efficacy, extensive treatment-emergent effects, and dose-limiting toxicities. The incidence of the latter two events may be associated with inhibition-driven pharmacology as a high and sustained concentration of inhibitor is required for therapeutic effect. The degradation of PRMT1 using a proteolysis targeting chimera (PROTAC) may be superior to inhibition as proceeds via event-driven pharmacology where a PROTAC acts catalytically at a low dose. PROTACs containing the same pharmacophore as GSK3368715, combined with a motif that recruits the VHL or CRBN E3-ligase, were synthesised. Suitable cell permeability and target engagement were shown for selected candidates by the detection of downstream effects of PRMT1 inhibition and by a NanoBRET assay for E3-ligase binding, however the candidates did not induce PRMT1 degradation. This paper is the first reported investigation of PRMT1 for targeted protein degradation and provides hypotheses and insights to assist the design of PROTACs for PRMT1 and other novel target proteins.
Collapse
Affiliation(s)
- Poppy L Martin
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, United Kingdom
| | | | - Shalini V Rao
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, CH2 ORE, United Kingdom
| | - Stephen J Walsh
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, United Kingdom
| | - Jason S Carroll
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, CH2 ORE, United Kingdom
| | - David R Spring
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, United Kingdom
| |
Collapse
|
37
|
Brodermann MH, Henderson EK, Sellar RS. The emerging role of targeted protein degradation to treat and study cancer. J Pathol 2024; 263:403-417. [PMID: 38886898 DOI: 10.1002/path.6301] [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: 01/18/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 06/20/2024]
Abstract
The evolution of cancer treatment has provided increasingly targeted strategies both in the upfront and relapsed disease settings. Small-molecule inhibitors and immunotherapy have risen to prominence with chimeric antigen receptor T-cells, checkpoint inhibitors, kinase inhibitors, and monoclonal antibody therapies being deployed across a range of solid organ and haematological malignancies. However, novel approaches are required to target transcription factors and oncogenic fusion proteins that are central to cancer biology and have generally eluded successful drug development. Thalidomide analogues causing protein degradation have been a cornerstone of treatment in multiple myeloma, but a lack of in-depth mechanistic understanding initially limited progress in the field. When the protein cereblon (CRBN) was found to mediate thalidomide analogues' action and CRBN's neo-targets were identified, existing and novel drug development accelerated, with applications outside multiple myeloma, including non-Hodgkin's lymphoma, myelodysplastic syndrome, and acute leukaemias. Critically, transcription factors were the first canonical targets described. In addition to broadening the application of protein-degrading drugs, resistance mechanisms are being overcome and targeted protein degradation is widening the scope of druggable proteins against which existing approaches have been ineffective. Examples of targeted protein degraders include molecular glues and proteolysis targeting chimeras (PROTACs): heterobifunctional molecules that bind to proteins of interest and cause proximity-induced ubiquitination and proteasomal degradation via a linked E3 ligase. Twenty years since their inception, PROTACs have begun progressing through clinical trials, with early success in targeting the oestrogen receptor and androgen receptor in breast and prostate cancer respectively. This review explores important developments in targeted protein degradation to both treat and study cancer. It also considers the potential advantages and challenges in the translational aspects of developing new treatments. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
| | - Elizabeth K Henderson
- Department of Haematology, UCL Cancer Institute, University College London, London, UK
| | - Rob S Sellar
- Department of Haematology, UCL Cancer Institute, University College London, London, UK
| |
Collapse
|
38
|
Xu M, Fu J, Pei Y, Li M, Kan W, Yan R, Xia C, Ma J, Wang P, Zhang Y, Gao Y, Yang Y, Zhou Y, Li J, Zhou B. Discovery of a Highly Potent, Selective and Efficacious USP7 Degrader for the Treatment of Acute Lymphoblastic Leukemia. J Med Chem 2024. [PMID: 39028938 DOI: 10.1021/acs.jmedchem.4c01134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
USP7 is an attractive therapeutic target for cancers, especially for acute lymphoblastic leukemia (ALL) with wild-type p53. Herein, we report the discovery of XM-U-14 as a highly potent, selective and efficacious USP7 proteolysis-targeting chimera degrader. XM-U-14 achieves DC50 values of 0.74 nM and Dmax of 93% in inducing USP7 degradation in RS4;11 cell lines, and also significantly inhibits ALL cell growth. XM-U-14 even at 5 mg/kg dosed daily effectively inhibits RS4;11 tumor growth with 64.7% tumor regressions and causes no signs of toxicity in mice. XM-U-14 is a promising USP7 degrader for further optimization for ALL treatment.
Collapse
Affiliation(s)
- Miaomiao Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Jingfeng Fu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Yuan Pei
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Mengna Li
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Weijuan Kan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Ruyu Yan
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Chaoyue Xia
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Jingkun Ma
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan, Guangdong 528400, China
| | - Peipei Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yan Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Yue Gao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yaxi Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Yubo Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan, Guangdong 528400, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jia Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan, Guangdong 528400, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Bing Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| |
Collapse
|
39
|
Duan H, Zhang J, Gui R, Lu Y, Pang A, Chen B, Shen L, Yu H, Li J, Xu T, Wang Y, Yao X, Zhang B, Lin N, Dong X, Zhou Y, Che J. Discovery of a Highly Potent and Selective BRD9 PROTAC Degrader Based on E3 Binder Investigation for the Treatment of Hematological Tumors. J Med Chem 2024; 67:11326-11353. [PMID: 38913763 DOI: 10.1021/acs.jmedchem.4c00883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
BRD9 is a pivotal epigenetic factor involved in cancers and inflammatory diseases. Still, the limited selectivity and poor phenotypic activity of targeted agents make it an atypically undruggable target. PROTAC offers an alternative strategy for overcoming the issue. In this study, we explored diverse E3 ligase ligands for the contribution of BRD9 PROTAC degradation. Through molecular docking, binding affinity analysis, and structure-activity relationship study, we identified a highly potent PROTAC E5, with excellent BRD9 degradation (DC50 = 16 pM) and antiproliferation in MV4-11 cells (IC50 = 0.27 nM) and OCI-LY10 cells (IC50 = 1.04 nM). E5 can selectively degrade BRD9 and induce cell cycle arrest and apoptosis. Moreover, the therapeutic efficacy of E5 was confirmed in xenograft tumor models, accompanied by further RNA-seq analysis. Therefore, these results may pave the way and provide the reference for the discovery and investigation of highly effective PROTAC degraders.
Collapse
Affiliation(s)
- Haiting Duan
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Jingyu Zhang
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Cancer Center of Zhejiang University, Hangzhou 310006, P. R. China
- Department of Clinical Pharmacology, Hangzhou Geriatric Hospital, Hangzhou, Zhejiang 310022, P. R. China
| | - Renzhao Gui
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan, Guangdong 528400, P. R. China
- School of Pharmacy, Zunyi Medical University, Zunyi 563000, P. R. China
| | - Yang Lu
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Ao Pang
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Beijing Chen
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan, Guangdong 528400, P. R. China
| | - Liteng Shen
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Hengyuan Yu
- State Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Jia Li
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan, Guangdong 528400, P. R. China
- School of Pharmacy, Zunyi Medical University, Zunyi 563000, P. R. China
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, P. R. China
| | - Tengfei Xu
- State Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Yuwei Wang
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712000, P. R. China
| | - Xiaojun Yao
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao 999078, P. R. China
| | - Bo Zhang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Cancer Center of Zhejiang University, Hangzhou 310006, P. R. China
- Department of Clinical Pharmacology, Hangzhou Geriatric Hospital, Hangzhou, Zhejiang 310022, P. R. China
| | - Nengming Lin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Cancer Center of Zhejiang University, Hangzhou 310006, P. R. China
- Department of Clinical Pharmacology, Hangzhou Geriatric Hospital, Hangzhou, Zhejiang 310022, P. R. China
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
- State Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Yubo Zhou
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan, Guangdong 528400, P. R. China
- School of Pharmacy, Zunyi Medical University, Zunyi 563000, P. R. China
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| | - Jinxin Che
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
- State Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| |
Collapse
|
40
|
Yan KN, Nie YQ, Wang JY, Yin GL, Liu Q, Hu H, Sun X, Chen XH. Accelerating PROTACs Discovery Through a Direct-to-Biology Platform Enabled by Modular Photoclick Chemistry. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400594. [PMID: 38689503 PMCID: PMC11234393 DOI: 10.1002/advs.202400594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/02/2024] [Indexed: 05/02/2024]
Abstract
Proteolysis targeting chimeras (PROTACs) have emerged as a promising strategy for drug discovery and exploring protein functions, offering a revolutionary therapeutic modality. Currently, the predominant approach to PROTACs discovery mainly relies on an empirical design-synthesis-evaluation process involving numerous cycles of labor-intensive synthesis-purification and bioassay data collection. Therefore, the development of innovative methods to expedite PROTAC synthesis and exploration of chemical space remains highly desired. Here, a direct-to-biology strategy is reported to streamline the synthesis of PROTAC libraries on plates, enabling the seamless transfer of reaction products to cell-based bioassays without the need for additional purification. By integrating amide coupling and light-induced primary amines and o-nitrobenzyl alcohols cyclization (PANAC) photoclick chemistry into a plate-based synthetic process, this strategy produces PROTAC libraries with high efficiency and structural diversity. Moreover, by employing this platform for PROTACs screening, we smoothly found potent PROTACs effectively inhibit triple-negative breast cancer (TNBC) cell growth and induce rapid, selective targeted degradation of cyclin-dependent kinase 9 (CDK9). The study introduces a versatile platform for assembling PROTACs on plates, followed by direct biological evaluation. This approach provides a promising opportunity for high-throughput synthesis of PROTAC libraries, thereby enhancing the efficiency of exploring chemical space and accelerating the discovery of PROTACs.
Collapse
Affiliation(s)
- Ke-Nian Yan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yong-Qiang Nie
- Jiangxi Key Laboratory of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Jia-Yu Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Guang-Liang Yin
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qia Liu
- University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Hao Hu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiaoxia Sun
- Jiangxi Key Laboratory of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Xiao-Hua Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| |
Collapse
|
41
|
Wang S, Feng Z, Qu C, Yu S, Zhang H, Deng R, Luo D, Pu C, Zhang Y, Li R. Novel Amphiphilic PROTAC with Enhanced Pharmacokinetic Properties for ALK Protein Degradation. J Med Chem 2024; 67:9842-9856. [PMID: 38839424 DOI: 10.1021/acs.jmedchem.3c02340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Advancements in anticancer strategies spotlight proteolysis targeting chimera (PROTAC) technology, yet it is hindered by poor water solubility and bioavailability. This study introduces a novel amphiphilic PROTAC, B1-PEG, synthesized through PEGylation of an optimized PROTAC molecule, B1, to enhance its properties. B1-PEG is engineered to self-organize into micelles in water and releases its active form in response to the tumor-specific high GSH environment. Comparative pharmacokinetic analysis revealed B1-PEG's superior bioavailability at 84.8%, outperforming the unmodified PROTAC molecule B1. When tested in a H3122 xenograft mouse model, B1-PEG significantly regressed tumors, underscoring its potential as a formidable candidate in targeted cancer therapy. Our findings offer a promising direction for overcoming bioavailability limitations in PROTAC drug design.
Collapse
Affiliation(s)
- Shirui Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhanzhan Feng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Can Qu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Su Yu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongjia Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Rui Deng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dan Luo
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu 610031, China
| | - Chunlan Pu
- Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu 610031, China
| | - Yan Zhang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Rui Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
42
|
Su LY, Tian Y, Zheng Q, Cao Y, Yao M, Wang S, Xu W, Xi C, Clocchiatti A, Nie G, Zhou H. Anti-tumor immunotherapy using engineered bacterial outer membrane vesicles fused to lysosome-targeting chimeras mediated by transferrin receptor. Cell Chem Biol 2024; 31:1219-1230.e5. [PMID: 38309277 DOI: 10.1016/j.chembiol.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/07/2023] [Accepted: 01/11/2024] [Indexed: 02/05/2024]
Abstract
The lysosome-targeting chimera (LYTAC) approach has shown promise for the targeted degradation of secreted and membrane proteins via lysosomes. However, there have been challenges in design, development, and targeting. Here, we have designed a genetically engineered transferrin receptor (TfR)-mediated lysosome-targeting chimera (TfR-LYTAC) that is efficiently internalized via TfR-mediate endocytosis and targets PD-L1 for lysosomal degradation in cultured cells but not in vivo due to short half-life and poor tumor targeting. A delivery platform was developed by fusing TfR-LYTAC to the surface of bacterial outer membrane vesicles (OMVs). The engineered OMV-LYTAC combines PD-1/PD-L1 pathway inhibition with LYTAC and immune activation by bacterial OMVs. OMV-LYTAC significantly reduced tumor growth in vivo. We have provided a modular and simple genetic strategy for lysosomal degradation as well as a delivery platform for in vivo tumor targeting. The study paves the way for the targeting and degradation of extracellular proteins using the TfR-LYTAC system.
Collapse
Affiliation(s)
- Ling-Yan Su
- College of Food Science and Technology, Yunnan Agricultural University, No. 452 Fengyuan Road, Kunming 650000, China; Yunnan Provincial Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650000, China
| | - Yang Tian
- College of Food Science and Technology, Yunnan Agricultural University, No. 452 Fengyuan Road, Kunming 650000, China; Yunnan Provincial Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650000, China
| | - Qiang Zheng
- Department of Cardiovascular Surgery, The First People Hospital of Yunnan Province, Xishan District, No.157 Jinbi Road, Kunming 650032, China
| | - Yu Cao
- Department of Cardiovascular Surgery, The First People Hospital of Yunnan Province, Xishan District, No.157 Jinbi Road, Kunming 650032, China
| | - Mengyu Yao
- Department of Cardiovascular Surgery, The First People Hospital of Yunnan Province, Xishan District, No.157 Jinbi Road, Kunming 650032, China
| | - Shuangping Wang
- College of Food Science and Technology, Yunnan Agricultural University, No. 452 Fengyuan Road, Kunming 650000, China
| | - Wen Xu
- College of Food Science and Technology, Yunnan Agricultural University, No. 452 Fengyuan Road, Kunming 650000, China
| | - Chuyu Xi
- College of Food Science and Technology, Yunnan Agricultural University, No. 452 Fengyuan Road, Kunming 650000, China
| | - Andrea Clocchiatti
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Hejiang Zhou
- College of Food Science and Technology, Yunnan Agricultural University, No. 452 Fengyuan Road, Kunming 650000, China; Yunnan Provincial Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650000, China.
| |
Collapse
|
43
|
Yao X, Mao J, Zhang H, Xiao Y, Wang Y, Liu H. Development of novel N-aryl-2,4-bithiazole-2-amine-based CYP1B1 degraders for reversing drug resistance. Eur J Med Chem 2024; 272:116488. [PMID: 38733885 DOI: 10.1016/j.ejmech.2024.116488] [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: 01/25/2024] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 05/13/2024]
Abstract
Extrahepatic cytochrome P450 1B1 (CYP1B1), which is highly expressed in non-small cell lung cancer, is an attractive target for cancer prevention, therapy, and overcoming drug resistance. Historically, CYP1B1 inhibition has been the primary therapeutic approach for treating CYP1B1-related malignancies, but its success has been limited. This study introduced CYP1B1 degradation as an alternative strategy to counter drug resistance and metastasis in CYP1B1-overexpressing non-small cell lung cancer A549/Taxol cells via a PROTAC strategy. Our investigation revealed that the identification of the potent CYP1B1 degrader PV2, achieving DC50 values of 1.0 nM and inducing >90 % CYP1B1 degradation at concentrations as low as 10 nM in A549/Taxol cells. Importantly, PV2 enhanced the sensitivity of the A549/Taxol subline to Taxol, possibly due to its stronger inhibitory effects on P-gp through CYP1B1 degradation. Additionally, compared to the CYP1B1 inhibitor A1, PV2 effectively suppressed the migration and invasion of A549/Taxol cells by inhibiting the FAK/SRC and EMT pathways. These findings hold promise for a novel therapy targeting advanced CYP1B1+ non-small cell lung cancer.
Collapse
Affiliation(s)
- Xiaoxuan Yao
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, PR China
| | - Jianping Mao
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, PR China
| | - Haoyu Zhang
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, PR China
| | - Yi Xiao
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, PR China
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, PR China.
| | - Hongzhuo Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, Liaoning, PR China.
| |
Collapse
|
44
|
Guan T, Zhang Y, Li S, Zhang W, Song Y, Li Y, He Y, Chen Y. Discovery of an efficacious KDM5B PROTAC degrader GT-653 up-regulating IFN response genes in prostate cancer. Eur J Med Chem 2024; 272:116494. [PMID: 38749268 DOI: 10.1016/j.ejmech.2024.116494] [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: 03/18/2024] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/27/2024]
Abstract
Epigenetic alterations promote cancer development by regulating the expression of various oncogenes and anti-oncogenes. Histone methylation modification represents a pivotal area in epigenetic research and numerous publications have demonstrated that aberrant histone methylation is highly correlated with tumorigenesis and development. As a key histone demethylase, lysine-specific demethylase 5B (KDM5B) demethylates lysine 4 of histone 3 (H3K4) and serves as a transcriptional repressor of certain tumor suppressor genes. Meanwhile, KDM5B inhibits STING-induced intrinsic immune response of tumor cells or recruits SETDB1 through non-enzymatic function to silence reverse transcription elements to promote immune escape. The conventional small molecule inhibitors can only inhibit the enzymatic function of KDM5B with no effect on the non-enzymatic function. In the article, we present the development of the first series of KDM5B degraders based on CPI-455 to inhibit the non-enzymatic function. Among them, GT-653 showed optimal KDM5B degradation efficiency in a ubiquitin proteasome-dependent manner. GT-653 efficiently reduced KDM5B protein levels without affecting KDM5B transcription. Interestingly, GT-653 increased H3K4me3 levels and activated the type-I interferon signaling pathway in 22RV1 cells without significant phenotypic response on cell proliferation.
Collapse
Affiliation(s)
- Tian Guan
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yingshuang Zhang
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Shen Li
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Wenbao Zhang
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yuxuan Song
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yuzhan Li
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yundong He
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Yihua Chen
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China; School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, 650500, China.
| |
Collapse
|
45
|
Tian Y, Seifermann M, Bauer L, Luchena C, Wiedmann JJ, Schmidt S, Geisel A, Afonin S, Höpfner J, Brehm M, Liu X, Hopf C, Popova AA, Levkin PA. High-Throughput Miniaturized Synthesis of PROTAC-Like Molecules. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307215. [PMID: 38258390 DOI: 10.1002/smll.202307215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/03/2024] [Indexed: 01/24/2024]
Abstract
The development of miniaturized high-throughput in situ screening platforms capable of handling the entire process of drug synthesis to final screening is essential for advancing drug discovery in the future. In this study, an approach based on combinatorial solid-phase synthesis, enabling the efficient synthesis of libraries of proteolysis targeting chimeras (PROTACs) in an array format is presented. This on-chip platform allows direct biological screening without the need for transfer steps. UV-induced release of target molecules into individual droplets facilitates further on-chip experimentation. Utilizing a mitogen-activated protein kinase kinases (MEK1/2) degrader as a template, a series of 132 novel PROTAC-like molecules is synthesized using solid-phase Ugi reaction. These compounds are further characterized using various methods, including matrix-assisted laser desorption ionization mass spectrometry (MALDI-MS) imaging, while consuming only a few milligrams of starting materials in total. Furthermore, the feasibility of culturing cancer cells on the modified spots and quantifying the effect of MEK suppression is demonstrated. The miniaturized synthesis platform lays a foundation for high-throughput in situ biological screening of potent PROTACs for potential anticancer activity and offers the potential for accelerating the drug discovery process by integrating miniaturized synthesis and biological steps on the same array.
Collapse
Affiliation(s)
- Ye Tian
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Hermann-von Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Wenhuaxi Road 44, Jinan, 250012, China
- Department of Immunology, Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Sciences, Shandong University, Wenhuaxi Road 44, Jinan, 250012, China
| | - Maximilian Seifermann
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Hermann-von Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Liana Bauer
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Hermann-von Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Charlotte Luchena
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Hermann-von Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Janne J Wiedmann
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Hermann-von Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Stefan Schmidt
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), University of Applied Sciences Mannheim, Paul-Wittsack-Straße 10, 68163, Mannheim, Germany
| | - Alexander Geisel
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), University of Applied Sciences Mannheim, Paul-Wittsack-Straße 10, 68163, Mannheim, Germany
| | - Sergii Afonin
- Institute of Biological Interfaces (IBG-2), Karlsruhe Institute of Technology (KIT), POB 3640, 76021, Karlsruhe, Germany
| | - Julius Höpfner
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Hermann-von Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Marius Brehm
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Hermann-von Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Wenhuaxi Road 44, Jinan, 250012, China
| | - Carsten Hopf
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), University of Applied Sciences Mannheim, Paul-Wittsack-Straße 10, 68163, Mannheim, Germany
- Medical Faculty, Heidelberg University, Im Neuenheimer Feld 280, 69117, Heidelberg, Germany
- Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Theodor Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Anna A Popova
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Hermann-von Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Pavel A Levkin
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Hermann-von Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
46
|
Bricelj A, Dora Ng YL, Gobec M, Kuchta R, Hu W, Javornik Š, Rožič M, Gütschow M, Zheng G, Krönke J, Steinebach C, Sosič I. Design, Synthesis, and Evaluation of BCL-2 Targeting PROTACs. Chemistry 2024:e202400430. [PMID: 38818652 DOI: 10.1002/chem.202400430] [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: 01/31/2024] [Revised: 05/26/2024] [Accepted: 05/28/2024] [Indexed: 06/01/2024]
Abstract
BCL-2, a member of the BCL-2 protein family, is an antiapoptotic factor that regulates the intrinsic pathway of apoptosis. Due to its aberrant activity, it is frequently implicated in haematopoietic cancers and represents an attractive target for the development of therapeutics that antagonize its activity. A selective BCL-2 inhibitor, venetoclax, was approved for treating chronic lymphocytic leukaemia, acute myeloid leukemia, and other haematologic malignancies, validating BCL-2 as an anticancer target. Since then, alternative therapeutic approaches to modulate the activity of BCL-2 have been explored, such as antibody-drug conjugates and proteolysis-targeting chimeras. Despite numerous research groups focusing on developing degraders of BCL-2 family member proteins, selective BCL-2 PROTACs remain elusive, as disclosed compounds only show dual BCL-xL/BCL-2 degradation. Herein, we report our efforts to develop BCL-2 degraders by incorporating two BCL-2 binding moieties into chimeric compounds that aim to hijack one of three E3 ligases: CRBN, VHL, and IAPs. Even though our project did not result in obtaining a potent and selective BCL-2 PROTAC, our research will aid in understanding the narrow chemical space of BCL-2 degraders.
Collapse
Affiliation(s)
- Aleša Bricelj
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, SI-1000, Ljubljana, Slovenia
| | - Yuen Lam Dora Ng
- Department of Hematology, Oncology and Cancer Immunology, Charité - Universitätsmedizin Berlin, Hindenburgdamm 30, D-12203, Berlin, Germany
| | - Martina Gobec
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, SI-1000, Ljubljana, Slovenia
| | - Robert Kuchta
- Pharmaceutical Institute, Department of Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, D-53121, Bonn, Germany
| | - Wanyi Hu
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, 32610, Gainesville, FL, USA
| | - Špela Javornik
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, SI-1000, Ljubljana, Slovenia
| | - Miha Rožič
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, SI-1000, Ljubljana, Slovenia
| | - Michael Gütschow
- Pharmaceutical Institute, Department of Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, D-53121, Bonn, Germany
| | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, 32610, Gainesville, FL, USA
| | - Jan Krönke
- Department of Hematology, Oncology and Cancer Immunology, Charité - Universitätsmedizin Berlin, Hindenburgdamm 30, D-12203, Berlin, Germany
| | - Christian Steinebach
- Pharmaceutical Institute, Department of Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, D-53121, Bonn, Germany
| | - Izidor Sosič
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, SI-1000, Ljubljana, Slovenia
| |
Collapse
|
47
|
Wang J, Wang Y, Yang F, Luo Q, Hou Z, Xing Y, Lu F, Li Z, Yin F. A Novel Lysosome Targeting Chimera for Targeted Protein Degradation via Split-and-Mix Strategy. ACS Chem Biol 2024; 19:1161-1168. [PMID: 38662199 DOI: 10.1021/acschembio.4c00092] [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: 04/26/2024]
Abstract
Targeted protein degradation is becoming more and more important in the field of drug development. Compared with proteasomal-based degraders, lysosomal-based degraders have a broader target spectrum of targets, which have been demonstrated to have great potential, especially in degrading undruggable proteins. Recently, we developed a programmable and facile screening PROTAC development platform based on peptide self-assembly termed split-and-mix PROTAC (SM-PROTAC). In this study, we applied this technology for the development of lysosome-based degraders, named a split-and-mix chaperone-mediated autophagy-based degrader (SM-CMAD). We successfully demonstrated SM-CMAD as a universal platform by degrading several targets, including ERα, AR, MEK1/2, and BCR-ABL. Different from other lysosomal-based degraders, SM-CMAD was capable of facile screening with programmable ligand ratios. We believe that our work will promote the development of other multifunctional molecules and clinical translation for lysosomal-based degraders.
Collapse
Affiliation(s)
- Jinpeng Wang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Yuechen Wang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | | | - Qinhong Luo
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Department of Pharmacy, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Zhanfeng Hou
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Yun Xing
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Fei Lu
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Zigang Li
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Pingshan Translational Medicine Center, Shenzhen Bay Laboratory, Shenzhen 518118, China
| | - Feng Yin
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Pingshan Translational Medicine Center, Shenzhen Bay Laboratory, Shenzhen 518118, China
| |
Collapse
|
48
|
Liu L, Parolia A, Liu Y, Hou C, He T, Qiao Y, Eyunni S, Luo J, Li C, Wang Y, Zhou F, Huang W, Ren X, Wang Z, Chinnaiyan AM, Ding K. Discovery of LLC0424 as a Potent and Selective in Vivo NSD2 PROTAC Degrader. J Med Chem 2024; 67:6938-6951. [PMID: 38687638 PMCID: PMC11094793 DOI: 10.1021/acs.jmedchem.3c01765] [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: 09/22/2023] [Revised: 04/13/2024] [Accepted: 04/19/2024] [Indexed: 05/02/2024]
Abstract
Nuclear receptor-binding SET domain-containing 2 (NSD2), a methyltransferase that primarily installs the dimethyl mark on lysine 36 of histone 3 (H3K36me2), has been recognized as a promising therapeutic target against cancer. However, existing NSD2 inhibitors suffer from low activity or inferior selectivity, and none of them can simultaneously remove the methyltransferase activity and chromatin binding function of NSD2. Herein we report the discovery of a novel NSD2 degrader LLC0424 by leveraging the proteolysis-targeting chimera technology. LLC0424 potently degraded NSD2 protein with a DC50 value of 20 nM and a Dmax value of 96% in acute lymphoblastic leukemia (ALL) RPMI-8402 cells. Mechanistic studies revealed LLC0424 to selectively induce NSD2 degradation in a cereblon- and proteasome-dependent fashion. LLC0424 also caused continuous downregulation of H3K36me2 and growth inhibition of ALL cell lines with NSD2 mutation. Importantly, intravenous or intraperitoneal injection of LLC0424 showed potent NSD2 degradation in vivo.
Collapse
Affiliation(s)
- Lianchao Liu
- State
Key Laboratory of Chemical Biology, Shanghai
Institute of Organic Chemistry, Chinese Academy of Sciences, no. 345 Lingling Road., Shanghai 200032, People’s Republic of China
| | - Abhijit Parolia
- 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
- Rogel
Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department
of Urology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yihan Liu
- Michigan
Center for Translational Pathology, University
of Michigan, Ann Arbor, Michigan 48109, United States
- Cancer
Biology
Program, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Caiyun Hou
- 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, People’s Republic of China
| | - Tongchen He
- Michigan
Center for Translational Pathology, University
of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yuanyuan Qiao
- 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
| | - Sanjana Eyunni
- 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
- Molecular
and Cellular Pathology Program, University
of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jie Luo
- 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
| | - Chungen Li
- State
Key Laboratory of Chemical Biology, Shanghai
Institute of Organic Chemistry, Chinese Academy of Sciences, no. 345 Lingling Road., Shanghai 200032, People’s Republic of China
| | - Yongxing Wang
- Livzon
Research Institute, Livzon Pharmaceutical
Group Inc., no. 38 Chuangye North Road, Jinwan District, Zhuhai 519000, China
| | - Fengtao 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, People’s Republic of China
| | - Weixue Huang
- State
Key Laboratory of Chemical Biology, Shanghai
Institute of Organic Chemistry, Chinese Academy of Sciences, no. 345 Lingling Road., Shanghai 200032, People’s Republic of China
| | - Xiaomei Ren
- State
Key Laboratory of Chemical Biology, Shanghai
Institute of Organic Chemistry, Chinese Academy of Sciences, no. 345 Lingling Road., Shanghai 200032, People’s Republic of China
| | - Zhen Wang
- State
Key Laboratory of Chemical Biology, Shanghai
Institute of Organic Chemistry, Chinese Academy of Sciences, no. 345 Lingling Road., Shanghai 200032, People’s Republic of China
| | - 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
- Rogel
Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department
of Urology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Howard
Hughes Medical Institute, 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, no. 345 Lingling Road., Shanghai 200032, People’s Republic of 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, People’s Republic of China
- Hangzhou Institute
of Medicine (HlM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
49
|
Chen J, Zhu W, Zhang W, Tong Y, Xu F, Pang J. Discovery of the First Potent DYRK2 Proteolysis Targeting Chimera Degraders. ACS Med Chem Lett 2024; 15:659-666. [PMID: 38746900 PMCID: PMC11089551 DOI: 10.1021/acsmedchemlett.4c00065] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 05/11/2025] Open
Abstract
Dual-specificity tyrosine phosphorylation-regulated kinase 2 (DYRK2) has been identified as a promising oncogenic driver of several types of cancer and is considered to be a critical cancer therapeutic target. Several inhibitors of DYRK2 have been reported, but no degraders have been found yet. In this work, we designed and synthesized the first series of proteolysis-targeting chimeras (PROTACs) using curcumin and its analogs as warheads to target and degrade DYRK2. The results of degradation assays showed that the compound CP134 could effectively downregulate the intracellular DYRK2 level (DC50 = 1.607 μM). Further mechanism of action experiments revealed that CP134 induced DYRK2 degradation through the ubiquitin-proteasome system. Altogether, CP134 disclosed in this study is the first potent DYRK2 degrader, which could serve as a valuable chemical tool for further evaluation of its therapeutic potential, and our results broaden the substrate spectrum of PROTAC-based degraders for further therapeutic applications.
Collapse
Affiliation(s)
- Jian Chen
- School
of Chemistry, Sun Yat-sen University, Guangzhou 510275, China
| | - Wentao Zhu
- School
of Chemistry, Sun Yat-sen University, Guangzhou 510275, China
| | - Wenqian Zhang
- School
of Chemistry, Sun Yat-sen University, Guangzhou 510275, China
| | - Yichen Tong
- School
of Chemistry, Sun Yat-sen University, Guangzhou 510275, China
| | - Fang Xu
- International
Cooperative Laboratory of Traditional Chinese Medicine Modernization
& Innovative Drug Development of Chinese Ministry of Education
(MOE) and Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Jiyan Pang
- School
of Chemistry, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
50
|
Mslati H, Gentile F, Pandey M, Ban F, Cherkasov A. PROTACable Is an Integrative Computational Pipeline of 3-D Modeling and Deep Learning To Automate the De Novo Design of PROTACs. J Chem Inf Model 2024; 64:3034-3046. [PMID: 38504115 DOI: 10.1021/acs.jcim.3c01878] [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: 03/21/2024]
Abstract
Proteolysis-targeting chimeras (PROTACs) that engage two biological targets at once are a promising technology in degrading clinically relevant protein targets. Since factors that influence the biological activities of PROTACs are more complex than those of a small molecule drug, we explored a combination of computational chemistry and deep learning strategies to forecast PROTAC activity and enable automated design. A new method named PROTACable was developed for the de novo design of PROTACs, which includes a robust 3-D modeling workflow to model PROTAC ternary complexes using a library of E3 ligase and linker and an SE(3)-equivariant graph transformer network to predict the activity of newly designed PROTACs. PROTACable is available at https://github.com/giaguaro/PROTACable/.
Collapse
Affiliation(s)
- Hazem Mslati
- Vancouver Prostate Centre, The University of British Columbia, Vancouver, British Columbia V6H 3Z6, Canada
| | - Francesco Gentile
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
- Ottawa Institute of Systems Biology, Ottawa, Ontario K1N 6N5, Canada
| | - Mohit Pandey
- Vancouver Prostate Centre, The University of British Columbia, Vancouver, British Columbia V6H 3Z6, Canada
| | - Fuqiang Ban
- Vancouver Prostate Centre, The University of British Columbia, Vancouver, British Columbia V6H 3Z6, Canada
| | - Artem Cherkasov
- Vancouver Prostate Centre, The University of British Columbia, Vancouver, British Columbia V6H 3Z6, Canada
| |
Collapse
|