1
|
Arkinson C, Dong KC, Gee CL, Martin A. Mechanisms and regulation of substrate degradation by the 26S proteasome. Nat Rev Mol Cell Biol 2025; 26:104-122. [PMID: 39362999 PMCID: PMC11772106 DOI: 10.1038/s41580-024-00778-0] [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] [Accepted: 08/23/2024] [Indexed: 10/05/2024]
Abstract
The 26S proteasome is involved in degrading and regulating the majority of proteins in eukaryotic cells, which requires a sophisticated balance of specificity and promiscuity. In this Review, we discuss the principles that underly substrate recognition and ATP-dependent degradation by the proteasome. We focus on recent insights into the mechanisms of conventional ubiquitin-dependent and ubiquitin-independent protein turnover, and discuss the plethora of modulators for proteasome function, including substrate-delivering cofactors, ubiquitin ligases and deubiquitinases that enable the targeting of a highly diverse substrate pool. Furthermore, we summarize recent progress in our understanding of substrate processing upstream of the 26S proteasome by the p97 protein unfoldase. The advances in our knowledge of proteasome structure, function and regulation also inform new strategies for specific inhibition or harnessing the degradation capabilities of the proteasome for the treatment of human diseases, for instance, by using proteolysis targeting chimera molecules or molecular glues.
Collapse
Affiliation(s)
- Connor Arkinson
- California Institute for Quantitative Biosciences, University of California at Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, CA, USA
| | - Ken C Dong
- Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, CA, USA
| | - Christine L Gee
- California Institute for Quantitative Biosciences, University of California at Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, CA, USA
| | - Andreas Martin
- California Institute for Quantitative Biosciences, University of California at Berkeley, Berkeley, CA, USA.
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA.
- Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, CA, USA.
| |
Collapse
|
2
|
Campos MA, Riha IA, Zhang C, Mozes C, Scheidt KA, Zhang X. Discovery of DCAF16 Binders for Targeted Protein Degradation. ACS Chem Biol 2025. [PMID: 39882752 DOI: 10.1021/acschembio.4c00799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Conventional small-molecule drugs primarily operate by inhibiting protein function, but this approach is limited when proteins lack well-defined ligand-binding pockets. Targeted protein degradation (TPD) offers an alternative approach by harnessing cellular degradation pathways to eliminate specific proteins. Recent studies have expanded the potential of TPD by identifying additional E3 ligases, with DCAF16 emerging as a promising candidate for facilitating protein degradation through both proteolysis-targeting chimera (PROTAC) and molecular glue mechanisms. In this study, we revisited a previously reported compound and discovered that it covalently binds to DCAF16. We further optimized it into a FKBP12-targeting PROTAC, MC-25B. This PROTAC engages DCAF16 at cysteines C177-179, leading to the degradation of nuclear-localized FKBP12. We further demonstrated the versatility of this DCAF16 recruiter by degrading additional endogenous proteins. Compared to the first-generation DCAF16-based PROTAC, which was derived from a fragment electrophile, this DCAF16 recruiter-based PROTAC exhibits improved proteome-wide selectivity.
Collapse
Affiliation(s)
- Miguel A Campos
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, United States
| | - Isabella A Riha
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Chenlu Zhang
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Chen Mozes
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Karl A Scheidt
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, United States
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Xiaoyu Zhang
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
- Center for Human Immunobiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| |
Collapse
|
3
|
Fan AT, Gadbois GE, Huang HT, Chaudhry C, Jiang J, Sigua LH, Smith ER, Wu S, Poirier GJ, Dunne-Dombrink K, Goyal P, Tao AJ, Sellers WR, Fischer ES, Donovan KA, Ferguson FM. A Kinetic Scout Approach Accelerates Targeted Protein Degrader Development. Angew Chem Int Ed Engl 2025; 64:e202417272. [PMID: 39602499 DOI: 10.1002/anie.202417272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 11/29/2024]
Abstract
Bifunctional molecules such as targeted protein degraders induce proximity to promote gain-of-function pharmacology. These powerful approaches have gained broad traction across academia and the pharmaceutical industry, leading to an intensive focus on strategies that can accelerate their identification and optimization. We and others have previously used chemical proteomics to map degradable target space, and these datasets have been used to develop and train multiparameter models to extend degradability predictions across the proteome. In this study, we now turn our attention to develop generalizable chemistry strategies to accelerate the development of new bifunctional degraders. We implement lysine-targeted reversible-covalent chemistry to rationally tune the binding kinetics at the protein-of-interest across a set of 25 targets. We define an unbiased workflow consisting of global proteomics analysis, IP/MS of ternary complexes and the E-STUB assay, to mechanistically characterize the effects of ligand residence time on targeted protein degradation and formulate hypotheses about the rate-limiting step of degradation for each target. Our key finding is that target residence time is a major determinant of degrader activity, and this can be rapidly and rationally tuned through the synthesis of a minimal number of analogues to accelerate early degrader discovery and optimization.
Collapse
Affiliation(s)
- Angela T Fan
- Department of Chemistry and Biochemistry, University of California, San Diego
| | - Gillian E Gadbois
- Department of Chemistry and Biochemistry, University of California, San Diego
| | - Hai-Tsang Huang
- The Broad Institute of Harvard and MIT
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston
| | - Charu Chaudhry
- Discovery Technologies Molecular Pharmacology, J&J Innovative Medicine, Spring House, Pennsylvania, 19477, United States
| | - Jiewei Jiang
- Department of Chemistry and Biochemistry, University of California, San Diego
| | - Logan H Sigua
- Medical Scientist Training Program, University of California, San Diego
| | - Emily R Smith
- Department of Chemistry and Biochemistry, University of California, San Diego
| | - Sitong Wu
- Department of Chemistry and Biochemistry, University of California, San Diego
| | - Grace J Poirier
- Department of Chemistry and Biochemistry, University of California, San Diego
| | - Kara Dunne-Dombrink
- Department of Chemistry and Biochemistry, University of California, San Diego
| | - Pavitra Goyal
- Department of Chemistry and Biochemistry, University of California, San Diego
| | - Andrew J Tao
- Department of Chemistry and Biochemistry, University of California, San Diego
| | - William R Sellers
- The Broad Institute of Harvard and MIT
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston
| | - Eric S Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston
| | - Katherine A Donovan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston
| | - Fleur M Ferguson
- Department of Chemistry and Biochemistry, University of California, San Diego
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego
| |
Collapse
|
4
|
Fu MJ, Jin H, Wang SP, Shen L, Liu HM, Liu Y, Zheng YC, Dai XJ. Unleashing the Power of Covalent Drugs for Protein Degradation. Med Res Rev 2025. [PMID: 39834319 DOI: 10.1002/med.22101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/28/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Targeted protein degradation (TPD) has emerged as a significant therapeutic approach for a variety of diseases, including cancer. Advances in TPD techniques, such as molecular glue (MG) and lysosome-dependent strategies, have shown substantial progress since the inception of the first PROTAC in 2001. The PROTAC methodology represents the forefront of TPD technology, with ongoing evaluation in more than 20 clinical trials for the treatment of diverse medical conditions. Two prominent PROTACs, ARV-471 and ARV-110, are currently undergoing phase III and II clinical trials, respectively. Traditional PROTACs are encountering obstacles such as limited binding affinity and a restricted range of E3 ligase ligands for facilitating the protein of interest (POI) degradation. Covalent medicines offer the potential to enhance PROTAC efficacy by enabling the targeting of previously considered "undruggable" shallow binding sites. Strategic alterations allow PROTAC to establish covalent connections with particular target proteins, including Kirsten rat sarcoma viral oncogene homolog (KRAS), Bruton's tyrosine kinase (BTK), epidermal growth factor receptor (EGFR), as well as E3 ligases such as DDB1 and CUL4 associated factor 16 (DCAF16) and Kelch-like ECH-associated protein 1 (Keap1). The concept of covalent degradation has also been utilized in various new forms of degraders, including covalent molecule glue (MG), in-cell click-formed proteolysis targeting chimera (CLIPTAC), HaloPROTAC, lysosome-targeting chimera (LYTAC) and GlueTAC. This review focuses on recent advancements in covalent degraders beyond covalent PROTACs and examines obstacles and future directions pertinent to this field.
Collapse
Affiliation(s)
- Meng-Jie Fu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Hang Jin
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Shao-Peng Wang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Liang Shen
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Hong-Min Liu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ying Liu
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi-Chao Zheng
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xing-Jie Dai
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardio-Cerebrovascular Drug, China Meheco Topfond Pharmaceutical Company, Zhumadian, Henan, China
| |
Collapse
|
5
|
Jochem M, Schrempf A, Wagner LM, Segal D, Cisneros J, Ng A, Winter GE, Krijgsveld J. Degradome analysis to identify direct protein substrates of small-molecule degraders. Cell Chem Biol 2025; 32:192-200.e6. [PMID: 39536762 DOI: 10.1016/j.chembiol.2024.10.007] [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: 02/09/2024] [Revised: 07/27/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
Targeted protein degradation (TPD) has emerged as a powerful strategy to selectively eliminate cellular proteins using small-molecule degraders, offering therapeutic promise for targeting proteins that are otherwise undruggable. However, a remaining challenge is to unambiguously identify primary TPD targets that are distinct from secondary downstream effects in the proteome. Here we introduce an approach for selective analysis of protein degradation by mass spectrometry (DegMS) at proteomic scale, which derives its specificity from the exclusion of confounding effects of altered transcription and translation induced by target depletion. We show that the approach efficiently operates at the timescale of TPD (hours) and we demonstrate its utility by analyzing the cyclin K degraders dCeMM2 and dCeMM4, which induce widespread transcriptional downregulation, and the GSPT1 degrader CC-885, an inhibitor of protein translation. Additionally, we apply DegMS to characterize a previously uncharacterized degrader, and identify the zinc-finger protein FIZ1 as a degraded target.
Collapse
Affiliation(s)
- Marco Jochem
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anna Schrempf
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | | | - Dmitri Segal
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Jose Cisneros
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Amanda Ng
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Georg E Winter
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Jeroen Krijgsveld
- German Cancer Research Center (DKFZ), Heidelberg, Germany; Heidelberg University, Medical Faculty, Heidelberg, Germany.
| |
Collapse
|
6
|
Lv MY, Hou DY, Liu SW, Cheng DB, Wang H. Strategy and Design of In Situ Activated Protein Hydrolysis Targeted Chimeras. ACS NANO 2025; 19:101-119. [PMID: 39731609 DOI: 10.1021/acsnano.4c11903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2024]
Abstract
Protein hydrolysis targeted chimeras (PROTACs) represent a different therapeutic approach, particularly relevant for overcoming challenges associated with traditional small molecule inhibitors. These challenges include targeting difficult proteins that are often deemed "undruggable" and addressing issues of acquired resistance. PROTACs employ the body's own E3 ubiquitin ligases to induce the degradation of specific proteins of interest (POIs) through the ubiquitin-proteasome pathway. This process is cyclical, allowing for broad applicability, potent protein degradation, and selective targeting. Despite their effectiveness, PROTACs can inadvertently target and degrade nonspecific proteins, potentially resulting in significant side effects and off-target toxicity. To address this concern, researchers have created stimuli-activated PROTACs that enhance targeted protein degradation while minimizing potential harm to healthy cells. These advanced PROTACs aim to improve the precision of degradation in both time and space. This article reviews the strategies for in situ activated PROTACs, highlighting key compounds and research advancements associated with various mechanisms of action. The insights presented here aim to guide further exploration in the field of activated PROTACs.
Collapse
Affiliation(s)
- Mei-Yu Lv
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Harbin 150001, China
| | - Da-Yong Hou
- Department of PET-CT/MRI, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Harbin 150001, China
| | - Shao-Wei Liu
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, P. R. China
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, P. R. China
| | - Haoran Wang
- Faculty of Materials Science, Shenzhen MSU-BIT University, Shenzhen 518100, P. R. China
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon 999077, Hong Kong, China
| |
Collapse
|
7
|
Wang Y, Hu X, Pandey S, Khatri U, Shen T, Subbiah V, Mooers BHM, Chao T, Wang S, Yu H, Sun X, Wu J, Cai J. Targeting Oncogenic RET Kinase by Simultaneously Inhibiting Kinase Activity and Degrading the Protein. J Med Chem 2025; 68:81-94. [PMID: 39723919 DOI: 10.1021/acs.jmedchem.4c01424] [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] [Indexed: 12/28/2024]
Abstract
The rearranged-during-transfection (RET) kinase is a validated target for the treatment of RET-altered cancers. Currently approved RET-selective kinase inhibitors, selpercatinib (LOXO-292) and pralsetinib (BLU-667), increase the oncogenic RET protein level upon treatment, which may affect their efficacy. We seek to reduce the oncogenic RET protein level and RET kinase activity simultaneously. Here, we report the development of proteolysis targeting chimera (PROTAC) degraders of oncogenic RET protein. Compound YW-N-7 exhibited dual action of selectively inhibiting and depleting RET protein both in vitro and in vivo. Proteomic analysis indicated that YW-N-7 is highly specific to RET. In cell cultures, reducing RET fusion protein potentiated the activity of LOXO-292. Furthermore, YW-N-7 showed significant activity in inhibiting KIF5B-RET-driven xenograft tumors in animals. This study exemplifies the feasibility of simultaneously inhibiting and degrading oncogenic RET kinase for cancer therapy.
Collapse
Affiliation(s)
- Yafeng Wang
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Xueqing Hu
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Shriya Pandey
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Ujjwol Khatri
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Tao Shen
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Vivek Subbiah
- Early-Phase Drug Development, Sarah Cannon Research Institute, Nashville, Tennessee 37203, United States
| | - Blaine H M Mooers
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Ting Chao
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Shaohui Wang
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, United States
| | - Huaxuan Yu
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Xingmin Sun
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, United States
| | - Jie Wu
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| |
Collapse
|
8
|
Liang J, Tian J, Zhang H, Li H, Chen L. Proteomics: An In-Depth Review on Recent Technical Advances and Their Applications in Biomedicine. Med Res Rev 2025. [PMID: 39789883 DOI: 10.1002/med.22098] [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: 05/13/2024] [Revised: 10/11/2024] [Accepted: 12/12/2024] [Indexed: 01/12/2025]
Abstract
Proteins hold pivotal importance since many diseases manifest changes in protein activity. Proteomics techniques provide a comprehensive exploration of protein structure, abundance, and function in biological samples, enabling the holistic characterization of overall changes in organisms. Nowadays, the breadth of emerging methodologies in proteomics is unprecedentedly vast, with constant optimization of technologies in sample processing, data collection, data analysis, and its scope of application is steadily transitioning from the bench to the clinic. Here, we offer an insightful review of the technical developments in proteomics and its applications in biomedicine over the past 5 years. We focus on its profound contributions in profiling disease spectra, discovering new biomarkers, identifying promising drug targets, deciphering alterations in protein conformation, and unearthing protein-protein interactions. Moreover, we summarize the cutting-edge technologies and potential breakthroughs in the proteomics pipeline and provide the principal challenges in proteomics. Based on these, we aspire to broaden the applicability of proteomics and inspire researchers to enhance our understanding of complex biological systems by utilizing such techniques.
Collapse
Affiliation(s)
- Jing Liang
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Jundan Tian
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Huadong Zhang
- College of Pharmacy, Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Hua Li
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
- College of Pharmacy, Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Lixia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
9
|
Tang J, Moorthy R, Hirsch LE, Demir Ö, Baker ZD, Naumann JA, Jones KFM, Grillo MJ, Haefner ES, Shi K, Levy MJ, Gupta HB, Aihara H, Harris RS, Amaro RE, Levinson NM, Harki DA. Targeting N-Myc in neuroblastoma with selective Aurora kinase A degraders. Cell Chem Biol 2025:S2451-9456(24)00516-6. [PMID: 39778578 DOI: 10.1016/j.chembiol.2024.12.006] [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: 05/27/2024] [Revised: 09/09/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025]
Abstract
The N-Myc transcription factor, encoded by MYCN, is a mechanistically validated, yet challenging, target for neuroblastoma (NB) therapy development. In normal neuronal progenitors, N-Myc undergoes rapid degradation, while, in MYCN-amplified NB cells, Aurora kinase A (Aurora-A) binds to and stabilizes N-Myc, resulting in elevated protein levels. Here, we demonstrate that targeted protein degradation of Aurora-A decreases N-Myc levels. A potent Aurora-A degrader, HLB-0532259 (compound 4), was developed from an Aurora-A-binding ligand that engages the Aurora-A/N-Myc complex. HLB-0532259 promotes the degradation of Aurora-A, which elicits concomitant N-Myc degradation, with nanomolar potency and excellent selectivity. HLB-0532259 surpasses the cellular efficacy of established allosteric Aurora-A inhibitors, exhibits favorable pharmacokinetic properties, and elicits tumor reduction in a murine xenograft NB model. This study broadly delineates a strategy for targeting "undruggable" proteins that are reliant on accessory proteins for cellular stabilization.
Collapse
Affiliation(s)
- Jian Tang
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ramkumar Moorthy
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Laura E Hirsch
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Özlem Demir
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Zachary D Baker
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jordan A Naumann
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Katherine F M Jones
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael J Grillo
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ella S Haefner
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ke Shi
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michaella J Levy
- KCAS Bioanalytical and Biomarker Services, Olathe, KS 66061, USA
| | - Harshita B Gupta
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Hideki Aihara
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Reuben S Harris
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Howard Hughes Medical Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Rommie E Amaro
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nicholas M Levinson
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel A Harki
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA; Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA; Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
10
|
Fu S, Chen Z, Luo Z, Nie M, Fu T, Zhou Y, Yang Q, Zhu F, Ni F. Chem(Pro)2: the atlas of chemoproteomic probes labelling human proteins. Nucleic Acids Res 2025; 53:D1651-D1662. [PMID: 39436046 PMCID: PMC11701659 DOI: 10.1093/nar/gkae943] [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: 08/15/2024] [Revised: 09/25/2024] [Accepted: 10/11/2024] [Indexed: 10/23/2024] Open
Abstract
Chemoproteomic probes (CPPs) have been widely considered as powerful molecular biological tools that enable the highly efficient discovery of both binding proteins and modes of action for the studied compounds. They have been successfully used to validate targets and identify binders. The design of CPP has been considered extremely challenging, which asks for the generalization using a large number of probe data. However, none of the existing databases gives such valuable data of CPPs. Herein, a database entitled 'Chem(Pro)2' was therefore developed to systematically describe the atlas of diverse types of CPPs labelling human protein in living cell/lysate. With the booming application of chemoproteomic technique and artificial intelligence in current chemical biology study, Chem(Pro)2 was expected to facilitate the AI-based learning of interacting pattern among molecules for discovering innovative targets and new drugs. Till now, Chem(Pro)2 has been open to all users without any login requirement at: https://idrblab.org/chemprosquare/.
Collapse
Affiliation(s)
- Songsen Fu
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
- LeadArt Biotechnologies Ltd., Ningbo 315201, China
| | - Zhen Chen
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Zhiming Luo
- LeadArt Biotechnologies Ltd., Ningbo 315201, China
| | - Meiyun Nie
- LeadArt Biotechnologies Ltd., Ningbo 315201, China
| | - Tingting Fu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Ying Zhou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Qingxia Yang
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Feng Ni
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
- LeadArt Biotechnologies Ltd., Ningbo 315201, China
| |
Collapse
|
11
|
Wu D, Sun Q, Tang H, Xiao H, Luo J, Ouyang L, Sun Q. Acquired resistance to tyrosine kinase targeted therapy: mechanism and tackling strategies. Drug Resist Updat 2025; 78:101176. [PMID: 39642660 DOI: 10.1016/j.drup.2024.101176] [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: 09/17/2024] [Revised: 11/21/2024] [Accepted: 11/23/2024] [Indexed: 12/09/2024]
Abstract
Over the past two decades, tyrosine kinase inhibitors (TKIs) have rapidly emerged as pivotal targeted agents, offering promising therapeutic prospects for patients. However, as the cornerstone of targeted therapies, an increasing number of TKIs have been found to develop acquired resistance during treatment, making the challenge of overcoming this resistance a primary focus of current research. This review comprehensively examines the evolution of TKIs from multiple perspectives, with particular emphasis on the mechanisms underlying acquired resistance, innovative drug design strategies, inherent challenges, and future directions.
Collapse
Affiliation(s)
- Defa Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Qian Sun
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China; West China Medical Publishers, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Haolin Tang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Huan Xiao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Jiaxiang Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China.
| | - Qiu Sun
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China; West China Medical Publishers, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
12
|
Riha IA, Campos MA, Jin X, Wang FY, Zhang C, Dunne SF, Cravatt BF, Zhang X. Exploiting the DCAF16-SPIN4 interaction to identify DCAF16 ligands for PROTAC development. RSC Med Chem 2024:d4md00681j. [PMID: 39691404 PMCID: PMC11647575 DOI: 10.1039/d4md00681j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 12/03/2024] [Indexed: 12/19/2024] Open
Abstract
Traditional small molecule drugs often target protein activity directly, but challenges arise when proteins lack suitable functional sites. An alternative approach is targeted protein degradation (TPD), which directs proteins to cellular machinery for proteolytic degradation. Recent studies have identified additional E3 ligases suitable for TPD, expanding the potential of this approach. Among these, DCAF16 has shown promise in facilitating protein degradation through both PROTAC and molecular glue mechanisms. In this study, we developed a homogeneous time resolved fluorescence (HTRF) assay to discover new DCAF16 binders. Using an in-house electrophile library, we identified two diastereomeric compounds, with one engaging DCAF16 at cysteines C177-179 and another reducing its expression. We demonstrated that the compound covalently engaging DCAF16 can be transformed into a PROTAC capable of degrading FKBP12.
Collapse
Affiliation(s)
- Isabella A Riha
- Department of Chemistry, Northwestern University Evanston IL 60208 USA
| | - Miguel A Campos
- Department of Chemistry, Northwestern University Evanston IL 60208 USA
- Chemistry of Life Processes Institute, Northwestern University Evanston IL 60208 USA
| | - Xiaokang Jin
- Department of Chemistry, Northwestern University Evanston IL 60208 USA
| | - Fiona Y Wang
- Department of Chemistry, Northwestern University Evanston IL 60208 USA
| | - Chenlu Zhang
- Department of Chemistry, Northwestern University Evanston IL 60208 USA
| | - Sara F Dunne
- High Throughput Analysis Laboratory, Northwestern University Evanston IL 60208 USA
| | - Benjamin F Cravatt
- Department of Chemistry, The Scripps Research Institute La Jolla CA 92037 USA
| | - Xiaoyu Zhang
- Department of Chemistry, Northwestern University Evanston IL 60208 USA
- Chemistry of Life Processes Institute, Northwestern University Evanston IL 60208 USA
- International Institute for Nanotechnology, Northwestern University Evanston IL 60208 USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine Chicago IL 60611 USA
- Center for Human Immunobiology, Northwestern University Feinberg School of Medicine Chicago IL 60611 USA
| |
Collapse
|
13
|
Adhikari B, Schneider K, Diebold M, Sotriffer C, Wolf E. Identification of suitable target/E3 ligase pairs for PROTAC development using a rapamycin-induced proximity assay (RiPA). eLife 2024; 13:RP98450. [PMID: 39641357 PMCID: PMC11623929 DOI: 10.7554/elife.98450] [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] [Indexed: 12/07/2024] Open
Abstract
The development of proteolysis targeting chimeras (PROTACs), which induce the degradation of target proteins by bringing them into proximity with cellular E3 ubiquitin ligases, has revolutionized drug development. While the human genome encodes more than 600 different E3 ligases, current PROTACs use only a handful of them, drastically limiting their full potential. Furthermore, many PROTAC development campaigns fail because the selected E3 ligase candidates are unable to induce degradation of the particular target of interest. As more and more ligands for novel E3 ligases are discovered, the chemical effort to identify the best E3 ligase for a given target is exploding. Therefore, a genetic system to identify degradation-causing E3 ligases and suitable target/E3 ligase pairs is urgently needed. Here, we used the well-established dimerization of the FKBP12 protein and FRB domain by rapamycin to bring the target protein WDR5 into proximity with candidate E3 ligases. Strikingly, this rapamycin-induced proximity assay (RiPA) revealed that VHL, but not Cereblon, is able to induce WDR5 degradation - a finding previously made by PROTACs, demonstrating its predictive power. By optimizing the steric arrangement of all components and fusing the target protein with a minimal luciferase, RiPA can identify the ideal E3 for any target protein of interest in living cells, significantly reducing and focusing the chemical effort in the early stages of PROTAC development.
Collapse
Affiliation(s)
| | | | - Mathias Diebold
- Institute of Biochemistry, University of KielKielGermany
- Institut für Pharmazie und Lebensmittelchemie, University of WürzburgWürzburgGermany
| | - Christoph Sotriffer
- Institut für Pharmazie und Lebensmittelchemie, University of WürzburgWürzburgGermany
| | - Elmar Wolf
- Institute of Biochemistry, University of KielKielGermany
| |
Collapse
|
14
|
Pravin N, Jóźwiak K. PROTAC unleashed: Unveiling the synthetic approaches and potential therapeutic applications. Eur J Med Chem 2024; 279:116837. [PMID: 39305635 DOI: 10.1016/j.ejmech.2024.116837] [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: 07/02/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 10/28/2024]
Abstract
Proteolysis-Targeting Chimeras (PROTACs) are a novel class of bifunctional small molecules that alter protein levels by targeted degradation. This innovative approach uses the ubiquitin-proteasome system to selectively eradicate disease-associated proteins, providing a novel therapeutic strategy for a wide spectrum of diseases. This review delineates detailed synthetic approaches involved in PROTAC building blocks, including the ligand and protein binding parts, linker attached structural components of PROTACs and the actual PROTAC molecules. Furthermore, the recent advancements in PROTAC-mediated degradation of specific oncogenic and other disease-associated proteins, such as those involved in neurodegenerative, antiviral, and autoimmune diseases, were also discussed. Additionally, we described the current landscape of PROTAC clinical trials and highlighted key studies that underscore the translational potential of this emerging therapeutic modality. These findings demonstrate the versatility of PROTACs in modulating the levels of key proteins involved in various severe diseases.
Collapse
Affiliation(s)
- Narayanaperumal Pravin
- Department of Biopharmacy, Medical University of Lublin, Ul.W.Chodzki 4a, 20-093 Lublin, Poland.
| | - Krzysztof Jóźwiak
- Department of Biopharmacy, Medical University of Lublin, Ul.W.Chodzki 4a, 20-093 Lublin, Poland.
| |
Collapse
|
15
|
Huber AD, Lin W, Poudel S, Miller DJ, Chen T. PROTAC-mediated activation, rather than degradation, of a nuclear receptor reveals complex ligand-receptor interaction network. Structure 2024; 32:2352-2363.e8. [PMID: 39389062 DOI: 10.1016/j.str.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/30/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024]
Abstract
Proteolysis-targeting chimeras (PROTACs) are heterobifunctional molecules containing a ligand for a protein of interest linked to an E3 ubiquitin ligase ligand that induce protein degradation through E3 recruitment to the target protein. Small changes in PROTAC linkers can have drastic consequences, including loss of degradation activity, but the structural mechanisms governing such changes are unclear. To study this phenomenon, we screened PROTACs of diverse targeting modalities and identified dTAG-13 as an activator of the xenobiotic-sensing pregnane X receptor (PXR), which promiscuously binds various ligands. Characterization of dTAG-13 analogs and precursors revealed interplay between the PXR-binding moiety, linker, and E3 ligand that altered PXR activity without inducing degradation. A crystal structure of PXR ligand binding domain bound to a precursor ligand showed ligand-induced binding pocket distortions and a linker-punctured tunnel to the protein exterior at a region incompatible with E3 complex formation, highlighting the effects of linker environment on PROTAC activity.
Collapse
Affiliation(s)
- Andrew D Huber
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 1000, Memphis, TN 38105-3678, USA
| | - Wenwei Lin
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 1000, Memphis, TN 38105-3678, USA
| | - Shyaron Poudel
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 1000, Memphis, TN 38105-3678, USA
| | - Darcie J Miller
- Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 1000, Memphis, TN 38105-3678, USA.
| |
Collapse
|
16
|
Basu AA, Zhang C, Rouhimoghadam M, Vasudevan A, Reitsma JM, Zhang X. Harnessing the FBXW7 somatic mutant R465C for targeted protein degradation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.03.626601. [PMID: 39677626 PMCID: PMC11643027 DOI: 10.1101/2024.12.03.626601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Targeted protein degradation (TPD) is a pharmacological strategy that eliminates specific proteins from cells by harnessing cellular proteolytic degradation machinery. In proteasome-dependent TPD, expanding the repertoire of E3 ligases compatible with this approach could enhance the applicability of this strategy across various biological contexts. In this study, we discovered that a somatic mutant of FBXW7, R465C, can be exploited by heterobifunctional compounds for targeted protein degradation. This work demonstrates the potential of utilizing mutant E3 ligases that occur exclusively in diseased cells for TPD applications.
Collapse
Affiliation(s)
- Ananya A. Basu
- Department of Chemistry, Northwestern University, Evanston, IL 60208
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208
| | - Chenlu Zhang
- Department of Chemistry, Northwestern University, Evanston, IL 60208
| | - Milad Rouhimoghadam
- Technology & Therapeutic Platforms, AbbVie Incorporated, North Chicago, IL 60064
| | - Anil Vasudevan
- Technology & Therapeutic Platforms, AbbVie Incorporated, North Chicago, IL 60064
| | - Justin M. Reitsma
- Technology & Therapeutic Platforms, AbbVie Incorporated, North Chicago, IL 60064
| | - Xiaoyu Zhang
- Department of Chemistry, Northwestern University, Evanston, IL 60208
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611
- Center for Human Immunobiology, Northwestern University, Chicago, IL 60611
- International Institute for Nanotechnology, Northwestern University, Evanston, IL 60208
| |
Collapse
|
17
|
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
|
18
|
Li YD, Ma MW, Hassan MM, Hunkeler M, Teng M, Puvar K, Rutter JC, Lumpkin RJ, Sandoval B, Jin CY, Schmoker AM, Ficarro SB, Cheong H, Metivier RJ, Wang MY, Xu S, Byun WS, Groendyke BJ, You I, Sigua LH, Tavares I, Zou C, Tsai JM, Park PMC, Yoon H, Majewski FC, Sperling HT, Marto JA, Qi J, Nowak RP, Donovan KA, Słabicki M, Gray NS, Fischer ES, Ebert BL. Template-assisted covalent modification underlies activity of covalent molecular glues. Nat Chem Biol 2024; 20:1640-1649. [PMID: 39075252 PMCID: PMC11582070 DOI: 10.1038/s41589-024-01668-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/05/2024] [Indexed: 07/31/2024]
Abstract
Molecular glues are proximity-inducing small molecules that have emerged as an attractive therapeutic approach. However, developing molecular glues remains challenging, requiring innovative mechanistic strategies to stabilize neoprotein interfaces and expedite discovery. Here we unveil a trans-labeling covalent molecular glue mechanism, termed 'template-assisted covalent modification'. We identified a new series of BRD4 molecular glue degraders that recruit CUL4DCAF16 ligase to the second bromodomain of BRD4 (BRD4BD2). Through comprehensive biochemical, structural and mutagenesis analyses, we elucidated how pre-existing structural complementarity between DCAF16 and BRD4BD2 serves as a template to optimally orient the degrader for covalent modification of DCAF16Cys58. This process stabilizes the formation of BRD4-degrader-DCAF16 ternary complex and facilitates BRD4 degradation. Supporting generalizability, we found that a subset of degraders also induces GAK-BRD4BD2 interaction through trans-labeling of GAK. Together, our work establishes 'template-assisted covalent modification' as a mechanism for covalent molecular glues, which opens a new path to proximity-driven pharmacology.
Collapse
Affiliation(s)
- Yen-Der Li
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michelle W Ma
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Muhammad Murtaza Hassan
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA, USA
| | - Moritz Hunkeler
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Mingxing Teng
- Center for Drug Discovery, Department of Pathology & Immunology, and Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Kedar Puvar
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Justine C Rutter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ryan J Lumpkin
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Brittany Sandoval
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Cyrus Y Jin
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Anna M Schmoker
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Scott B Ficarro
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Blais Proteomics Center and Center for Emergent Drug Targets, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Hakyung Cheong
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rebecca J Metivier
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michelle Y Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shawn Xu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Woong Sub Byun
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA, USA
| | - Brian J Groendyke
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Inchul You
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA, USA
| | - Logan H Sigua
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Isidoro Tavares
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Blais Proteomics Center and Center for Emergent Drug Targets, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Charles Zou
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jonathan M Tsai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Paul M C Park
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hojong Yoon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Felix C Majewski
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA, USA
| | - Haniya T Sperling
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jarrod A Marto
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Blais Proteomics Center and Center for Emergent Drug Targets, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Jun Qi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Radosław P Nowak
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Katherine A Donovan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Mikołaj Słabicki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nathanael S Gray
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA, USA.
| | - Eric S Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
19
|
Vikal A, Maurya R, Patel BB, Sharma R, Patel P, Patil UK, Das Kurmi B. Protacs in cancer therapy: mechanisms, design, clinical trials, and future directions. Drug Deliv Transl Res 2024:10.1007/s13346-024-01754-z. [PMID: 39614036 DOI: 10.1007/s13346-024-01754-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2024] [Indexed: 12/01/2024]
Abstract
Cancer develops as a result of changes in both genetic and epigenetic mechanisms, which lead to the activation of oncogenes and the suppression of tumor suppressor genes. Despite advancements in cancer treatments, the primary approach still involves a combination of chemotherapy, radiotherapy, and surgery, typically providing a median survival of approximately five years for patients. Unfortunately, these therapeutic interventions often bring about substantial side effects and toxicities, significantly impacting the overall quality of life for individuals undergoing treatment. Therefore, urgent need of research required which comes up with effective treatment of cancer. This review explores the transformative role of Proteolysis-Targeting Chimeras (PROTACs) in cancer therapy. PROTACs, an innovative drug development strategy, utilize the cell's protein degradation machinery to selectively eliminate disease-causing proteins. The review covers the historical background, mechanism of action, design, and structure of PROTACs, emphasizing their precision in targeting oncogenic proteins. The discussion extends to the challenges, nanotechnology applications, and ongoing clinical trials, showcasing promising results and clinical progress. The review concludes with insights into patents, future directions, and the potential impact of PROTACs in addressing dysregulated protein expression across various diseases. Overall, it provides a concise yet comprehensive overview for researchers, clinicians, and industry professionals involved in developing targeted therapies.
Collapse
Affiliation(s)
- Akash Vikal
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Rashmi Maurya
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Brij Bihari Patel
- Department of Respiratory Medicine, School of Excellence in Pulmonary Medicines, Netaji Subhash Chandra Bose Medical College, Jabalpur, 482003, Madhya Pradesh, India
| | - Rajeev Sharma
- Department of Pharmacy, Amity University, Gwalior, 474005, Madhya Pradesh, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Umesh K Patil
- Department of Pharmaceutical Sciences, Dr. Hari Singh Gour University, Sagar, 470003, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India.
| |
Collapse
|
20
|
Pliatsika D, Blatter C, Riedl R. Targeted protein degradation: current molecular targets, localization, and strategies. Drug Discov Today 2024; 29:104178. [PMID: 39276920 DOI: 10.1016/j.drudis.2024.104178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/23/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
Targeted protein degradation (TPD) has revolutionized drug discovery by selectively eliminating specific proteins within and outside the cellular context. Over the past two decades, TPD has expanded its focus beyond well-established targets, exploring diverse proteins beyond cancer-related ones. This evolution extends the potential of TPD to various diseases. Notably, TPD can target proteins at demanding locations, such as the extracellular matrix (ECM) and cellular membranes, presenting both opportunities and challenges for future research. In this review, we comprehensively examine the exciting opportunities in the burgeoning field of TPD, highlighting different targets, their cellular environment, and innovative strategies for modern drug discovery.
Collapse
Affiliation(s)
- Dimanthi Pliatsika
- Institute of Chemistry and Biotechnology, Competence Center for Drug Discovery, Zurich University of Applied Sciences, CH-8820 Wädenswil, Switzerland
| | - Cindy Blatter
- Institute of Chemistry and Biotechnology, Competence Center for Drug Discovery, Zurich University of Applied Sciences, CH-8820 Wädenswil, Switzerland
| | - Rainer Riedl
- Institute of Chemistry and Biotechnology, Competence Center for Drug Discovery, Zurich University of Applied Sciences, CH-8820 Wädenswil, Switzerland.
| |
Collapse
|
21
|
Winter GE. Extrapolating Lessons from Targeted Protein Degradation to Other Proximity-Inducing Drugs. ACS Chem Biol 2024; 19:2089-2102. [PMID: 39264973 PMCID: PMC11494510 DOI: 10.1021/acschembio.4c00191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/14/2024]
Abstract
Targeted protein degradation (TPD) is an emerging pharmacologic strategy. It relies on small-molecule "degraders" that induce proximity of a component of an E3 ubiquitin ligase complex and a target protein to induce target ubiquitination and subsequent proteasomal degradation. Essentially, degraders thus expand the function of E3 ligases, allowing them to degrade proteins they would not recognize in the absence of the small molecule. Over the past decade, insights gained from identifying, designing, and characterizing various degraders have significantly enhanced our understanding of TPD mechanisms, precipitating in rational degrader discovery strategies. In this Account, I aim to explore how these insights can be extrapolated to anticipate both opportunities and challenges of utilizing the overarching concept of proximity-inducing pharmacology to manipulate other cellular circuits for the dissection of biological mechanisms and for therapeutic purposes.
Collapse
Affiliation(s)
- Georg E. Winter
- CeMM Research Center for
Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| |
Collapse
|
22
|
Wilms G, Schofield K, Maddern S, Foley C, Shaw Y, Smith B, Basantes LE, Schwandt K, Babendreyer A, Chavez T, McKee N, Gokhale V, Kallabis S, Meissner F, Rokey SN, Dunckley T, Montfort WR, Becker W, Hulme C. Discovery and Functional Characterization of a Potent, Selective, and Metabolically Stable PROTAC of the Protein Kinases DYRK1A and DYRK1B. J Med Chem 2024; 67:17259-17289. [PMID: 39344427 DOI: 10.1021/acs.jmedchem.4c01130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Small-molecule-induced protein degradation has emerged as a promising pharmacological modality for inactivating disease-relevant protein kinases. DYRK1A and DYRK1B are closely related protein kinases that are involved in pathological processes such as neurodegeneration, cancer development, and adaptive immune homeostasis. Herein, we report the development of the first DYRK1 proteolysis targeting chimeras (PROTACs) that combine a new ATP-competitive DYRK1 inhibitor with ligands for the E3 ubiquitin ligase component cereblon (CRBN) to induce ubiquitination and subsequent proteasomal degradation of DYRK1A and DYRK1B. The lead compound (DYR684) promoted fast, efficient, potent, and selective degradation of DYRK1A in cell-based assays. Interestingly, an enzymatically inactive splicing variant of DYRK1B (p65) resisted degradation. Compared to competitive kinase inhibition, targeted degradation of DYRK1 by DYR684 provided improved suppression of downstream signaling. Collectively, our results identify DYRKs as viable targets for PROTAC-mediated degradation and qualify DYR684 as a useful chemical probe for DYRK1A and DYRK1B.
Collapse
Affiliation(s)
- Gerrit Wilms
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany
| | - Kevin Schofield
- Division of Drug Discovery and Development, Department of Pharmacology and Toxicology, College of Pharmacy The University of Arizona, Tucson, Arizona 85721, United States
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Shayna Maddern
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Christopher Foley
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Yeng Shaw
- Division of Drug Discovery and Development, Department of Pharmacology and Toxicology, College of Pharmacy The University of Arizona, Tucson, Arizona 85721, United States
| | - Breland Smith
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - L Emilia Basantes
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Katharina Schwandt
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany
| | - Aaron Babendreyer
- Institute of Molecular Pharmacology, RWTH Aachen University, Aachen 52074, Germany
| | - Timothy Chavez
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Nicholas McKee
- Division of Drug Discovery and Development, Department of Pharmacology and Toxicology, College of Pharmacy The University of Arizona, Tucson, Arizona 85721, United States
| | - Vijay Gokhale
- BIO5 Institute, The University of Arizona, Tucson, Arizona 85721, United States
| | - Sebastian Kallabis
- Core Facility Translational Proteomics, Institute of Innate Immunity, University Hospital Bonn, Bonn 53127, Germany
| | - Felix Meissner
- Department of Systems Immunology and Proteomics, Institute of Innate Immunity, University Hospital Bonn, Bonn 53127, Germany
| | - Samantha N Rokey
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Travis Dunckley
- ASU-Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, Arizona 85281, United States
| | - William R Montfort
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Walter Becker
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany
| | - Christopher Hulme
- Division of Drug Discovery and Development, Department of Pharmacology and Toxicology, College of Pharmacy The University of Arizona, Tucson, Arizona 85721, United States
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
23
|
Mi D, Huang A, Song J, Chen Y. A long march for targeted protein degradation in the new era: expanding E3 ligases. Future Med Chem 2024; 16:2189-2191. [PMID: 39373160 PMCID: PMC11622777 DOI: 10.1080/17568919.2024.2409618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/17/2024] [Indexed: 10/08/2024] Open
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
| | - Anxin Huang
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jingfeng Song
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, 650500, China
- Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, Yunnan, 650500, 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
- Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, Yunnan, 650500, China
| |
Collapse
|
24
|
Baek K, Metivier RJ, Roy Burman SS, Bushman JW, Yoon H, Lumpkin RJ, Abeja DM, Lakshminarayan M, Yue H, Ojeda S, Verano AL, Gray NS, Donovan KA, Fischer ES. Unveiling the hidden interactome of CRBN molecular glues with chemoproteomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612438. [PMID: 39314457 PMCID: PMC11419069 DOI: 10.1101/2024.09.11.612438] [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: 09/25/2024]
Abstract
Targeted protein degradation and induced proximity refer to strategies that leverage the recruitment of proteins to facilitate their modification, regulation or degradation. As prospective design of glues remains challenging, unbiased discovery methods are needed to unveil hidden chemical targets. Here we establish a high throughput affinity purification mass spectrometry workflow in cell lysates for the unbiased identification of molecular glue targets. By mapping the targets of 20 CRBN-binding molecular glues, we identify 298 protein targets and demonstrate the utility of enrichment methods for identifying novel targets overlooked using established methods. We use a computational workflow to estimate target confidence and perform a biochemical screen to identify a lead compound for the new non-ZF target PPIL4. Our study provides a comprehensive inventory of targets chemically recruited to CRBN and delivers a robust and scalable workflow for identifying new drug-induced protein interactions in cell lysates.
Collapse
Affiliation(s)
- Kheewoong Baek
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Rebecca J. Metivier
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Shourya S. Roy Burman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Jonathan W. Bushman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Hojong Yoon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ryan J. Lumpkin
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Dinah M. Abeja
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Megha Lakshminarayan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Hong Yue
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Samuel Ojeda
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Alyssa L. Verano
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Nathanael S. Gray
- Department of Chemical and Systems Biology, ChEM-H and Stanford Cancer Institute, Stanford Medical School, Stanford University, Stanford, CA, 94305, USA
| | - Katherine A. Donovan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Eric S. Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
25
|
Chen Y, Xia Z, Suwal U, Rappu P, Heino J, De Wever O, De Geest BG. Dual-ligand PROTACS mediate superior target protein degradation in vitro and therapeutic efficacy in vivo. Chem Sci 2024:d4sc03555k. [PMID: 39391379 PMCID: PMC11462456 DOI: 10.1039/d4sc03555k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024] Open
Abstract
Proteolysis targeting chimeras (PROTACs) are revolutionizing the drug development landscape due to their unique ability to selectively degrade disease-associated proteins. Conventional PROTACs are bivalent entities that induce ubiquitination and subsequent proteolysis of a chosen protein of interest (POI) by forming a ternary complex with an E3 ligase. We hypothesized that dual-ligand PROTACs, featuring two copies each of a POI ligand and an E3 ligase ligand, would facilitate the formation of high-avidity, long-lived ternary complexes inside cells, thereby increasing POI degradation potency. To this end, we developed a convergent synthesis route, using l-aspartic acid as a building block for homodimer synthesis, followed by copper-catalyzed azide-alkyne cycloaddition (CuAAC) to conjugate both dimers through a flexible linker. Dual-ligand PROTACs achieved up to a tenfold increase in degradation efficiency and a hundredfold increase in cytotoxicity in vitro across various cancer cell lines compared to their single-ligand counterparts. Furthermore, dual-ligand PROTACs sustain prolonged protein degradation, up to 60 hours after pulsing and washout. In vivo, in a mouse tumor model, the superior therapeutic activity of dual ligand PROTACs was observed.
Collapse
Affiliation(s)
- Yong Chen
- Department of Pharmaceutics, Ghent University 9000 Ghent Belgium
- Cancer Research Institute Ghent 9000 Ghent Belgium
| | - Zihan Xia
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University 9000 Ghent Belgium
- Cancer Research Institute Ghent 9000 Ghent Belgium
| | - Ujjwal Suwal
- Department of Life Technologies, InFLAMES Flagship, University of Turku 20520 Turku Finland
| | - Pekka Rappu
- Department of Life Technologies, InFLAMES Flagship, University of Turku 20520 Turku Finland
| | - Jyrki Heino
- Department of Life Technologies, InFLAMES Flagship, University of Turku 20520 Turku Finland
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University 9000 Ghent Belgium
- Cancer Research Institute Ghent 9000 Ghent Belgium
| | - Bruno G De Geest
- Department of Pharmaceutics, Ghent University 9000 Ghent Belgium
- Cancer Research Institute Ghent 9000 Ghent Belgium
| |
Collapse
|
26
|
Li Z, Liu H, He Z, Chakravarty A, Golden RP, Jiang Z, You I, Yue H, Donovan KA, Du G, Che J, Tse J, Che I, Lu W, Fischer ES, Zhang T, Gray NS, Yang PL. Discovery of Potent Degraders of the Dengue Virus Envelope Protein. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405829. [PMID: 39145423 PMCID: PMC11516100 DOI: 10.1002/advs.202405829] [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: 05/28/2024] [Revised: 06/26/2024] [Indexed: 08/16/2024]
Abstract
Targeted protein degradation has been widely adopted as a new approach to eliminate both established and previously recalcitrant therapeutic targets. Here, it is reported that the development of small molecule degraders of the envelope (E) protein of dengue virus. Two classes of bivalent E-degraders are developed by linking two previously reported E-binding small molecules, GNF-2, and CVM-2-12-2, to a glutarimide-based recruiter of the CRL4CRBN ligase to effect proteosome-mediated degradation of the E protein. ZXH-2-107 (based on GNF-2) is an E-degrader with ABL inhibitory activity while ZXH-8-004 (based on CVM-2-12-2) is a selective and potent E-degrader. These two compounds provide proof of concept that difficult-to-drug targets such as a viral envelope protein can be effectively eliminated using a bivalent degrader and provide starting points for the future development of a new class of direct-acting antiviral drugs.
Collapse
Affiliation(s)
- Zhengnian Li
- Department of Chemical and Systems BiologyChem‐H and Stanford Cancer InstituteStanford MedicineStanford University290 Jane Stanford WayStanfordCA94305USA
| | - Han‐Yuan Liu
- Department of Microbiology and ImmunologyStanford University School of Medicine279 Campus DrivePalo AltoCA94305USA
| | - Zhixiang He
- Department of Cancer BiologyDana‐Farber Cancer Institute450 Brookline AvenueBoston02215USA
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical School240 Longwood AvenueBoston02115USA
| | - Antara Chakravarty
- Department of Microbiology and ImmunologyStanford University School of Medicine279 Campus DrivePalo AltoCA94305USA
| | - Ryan P. Golden
- Department of Chemical and Systems BiologyChem‐H and Stanford Cancer InstituteStanford MedicineStanford University290 Jane Stanford WayStanfordCA94305USA
| | - Zixuan Jiang
- Department of Chemical and Systems BiologyChem‐H and Stanford Cancer InstituteStanford MedicineStanford University290 Jane Stanford WayStanfordCA94305USA
| | - Inchul You
- Department of Chemical and Systems BiologyChem‐H and Stanford Cancer InstituteStanford MedicineStanford University290 Jane Stanford WayStanfordCA94305USA
| | | | - Katherine A. Donovan
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical School240 Longwood AvenueBoston02115USA
| | - Guangyan Du
- Department of Cancer BiologyDana‐Farber Cancer Institute450 Brookline AvenueBoston02215USA
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical School240 Longwood AvenueBoston02115USA
| | - Jianwei Che
- Department of Cancer BiologyDana‐Farber Cancer Institute450 Brookline AvenueBoston02215USA
| | - Jason Tse
- Department of Chemical and Systems BiologyChem‐H and Stanford Cancer InstituteStanford MedicineStanford University290 Jane Stanford WayStanfordCA94305USA
| | - Isaac Che
- Department of Chemical and Systems BiologyChem‐H and Stanford Cancer InstituteStanford MedicineStanford University290 Jane Stanford WayStanfordCA94305USA
| | - Wenchao Lu
- Department of Chemical and Systems BiologyChem‐H and Stanford Cancer InstituteStanford MedicineStanford University290 Jane Stanford WayStanfordCA94305USA
| | - Eric S. Fischer
- Department of Cancer BiologyDana‐Farber Cancer Institute450 Brookline AvenueBoston02215USA
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical School240 Longwood AvenueBoston02115USA
| | - Tinghu Zhang
- Department of Chemical and Systems BiologyChem‐H and Stanford Cancer InstituteStanford MedicineStanford University290 Jane Stanford WayStanfordCA94305USA
| | - Nathanael S. Gray
- Department of Chemical and Systems BiologyChem‐H and Stanford Cancer InstituteStanford MedicineStanford University290 Jane Stanford WayStanfordCA94305USA
| | - Priscilla L. Yang
- Department of Microbiology and ImmunologyStanford University School of Medicine279 Campus DrivePalo AltoCA94305USA
| |
Collapse
|
27
|
Goebel GL, Giannino N, Lampe P, Qiu X, Schloßhauer JL, Imig J, Sievers S, Wu P. Profiling Cellular Morphological Changes Induced by Dual-Targeting PROTACs of Aurora Kinase and RNA-Binding Protein YTHDF2. Chembiochem 2024; 25:e202400183. [PMID: 38837838 DOI: 10.1002/cbic.202400183] [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: 02/28/2024] [Revised: 06/02/2024] [Accepted: 06/04/2024] [Indexed: 06/07/2024]
Abstract
Proteolysis targeting chimeras (PROTACs) are new chemical modalities that degrade proteins of interest, including established kinase targets and emerging RNA-binding proteins (RBPs). Whereas diverse sets of biochemical, biophysical and cellular assays are available for the evaluation and optimizations of PROTACs in understanding the involved ubiquitin-proteasome-mediated degradation mechanism and the structure-degradation relationship, a phenotypic method profiling the cellular morphological changes is rarely used. In this study, first, we reported the only examples of PROTACs degrading the mRNA-binding protein YTHDF2 via screening of multikinase PROTACs. Second, we reported the profiling of cellular morphological changes of the dual kinase- and RBP-targeting PROTACs using the unbiased cell painting assay (CPA). The CPA analysis revealed the high biosimilarity with the established aurora kinase cluster and annotated aurora kinase inhibitors, which reflected the association between YTHDF2 and the aurora kinase signaling network. Broadly, the results demonstrated that the cell painting assay can be a straightforward and powerful approach to evaluate PROTACs. Complementary to the existing biochemical, biophysical and cellular assays, CPA provided a new perspective in characterizing PROTACs at the cellular morphology.
Collapse
Affiliation(s)
- Georg L Goebel
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Otto-Hahn Str. 11, Dortmund, 44227, Germany
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Str. 11, Dortmund, 44227, Germany
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn Str. 6, Dortmund, 44227, Germany
| | - Nicole Giannino
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Otto-Hahn Str. 11, Dortmund, 44227, Germany
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Str. 11, Dortmund, 44227, Germany
| | - Philipp Lampe
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Str. 11, Dortmund, 44227, Germany
- Compound Management and Screening Center, Otto-Hahn Str. 15, Dortmund, 44227, Germany
| | - Xiaqiu Qiu
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Otto-Hahn Str. 11, Dortmund, 44227, Germany
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Str. 11, Dortmund, 44227, Germany
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn Str. 6, Dortmund, 44227, Germany
| | - Jeffrey L Schloßhauer
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Otto-Hahn Str. 11, Dortmund, 44227, Germany
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Str. 11, Dortmund, 44227, Germany
| | - Jochen Imig
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Otto-Hahn Str. 11, Dortmund, 44227, Germany
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Str. 11, Dortmund, 44227, Germany
| | - Sonja Sievers
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Str. 11, Dortmund, 44227, Germany
- Compound Management and Screening Center, Otto-Hahn Str. 15, Dortmund, 44227, Germany
| | - Peng Wu
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Otto-Hahn Str. 11, Dortmund, 44227, Germany
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Str. 11, Dortmund, 44227, Germany
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn Str. 6, Dortmund, 44227, Germany
| |
Collapse
|
28
|
Zhuang Z, Byun WS, Kozicka Z, Dwyer BG, Donovan KA, Jiang Z, Jones HM, Abeja DM, Nix MN, Zhong J, Słabicki M, Fischer ES, Ebert BL, Gray NS. Discovery of electrophilic degraders that exploit S NAr chemistry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.615094. [PMID: 39386645 PMCID: PMC11463635 DOI: 10.1101/2024.09.25.615094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Targeted covalent inhibition (TCI) and targeted protein degradation (TPD) have proven effective in pharmacologically addressing formerly 'undruggable' targets. Integration of both methodologies has resulted in the development of electrophilic degraders where recruitment of a suitable E3 ubiquitin ligase is achieved through formation of a covalent bond with a cysteine nucleophile. Expanding the scope of electrophilic degraders requires the development of electrophiles with tempered reactivity that enable selective ligase recruitment and reduce cross-reactivity with other cellular nucleophiles. In this study, we report the use of chemical moieties that enable nucleophilic aromatic substitution (SNAr) reactions in the rational design of electrophilic protein degraders. Appending an SNAr covalent warhead to several preexisting small molecule inhibitors transformed them into degraders, obviating the need for a defined E3 ligase recruiter. The SNAr covalent warhead is versatile; it can recruit various E3 ligases, including DDB1 and CUL4 associated factor 11 (DCAF11), DDB1 and CUL4 associated factor 16 (DCAF16), and possibly others. The incorporation of an SNAr covalent warhead into the BRD4 inhibitor led to the discovery of degraders with low picomolar degradation potency. Furthermore, we demonstrate the broad applicability of this approach through rational functional switching from kinase inhibitors into potent degraders.
Collapse
Affiliation(s)
- Zhe Zhuang
- Department of Chemical and Systems Biology, ChEM-H, and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA, USA
- These authors contributed equally: Zhe Zhuang, Woong Sub Byun
| | - Woong Sub Byun
- Department of Chemical and Systems Biology, ChEM-H, and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA, USA
- These authors contributed equally: Zhe Zhuang, Woong Sub Byun
| | - Zuzanna Kozicka
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Brendan G. Dwyer
- Department of Chemical and Systems Biology, ChEM-H, and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA, USA
| | - Katherine A. Donovan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Zixuan Jiang
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Hannah M. Jones
- Department of Chemical and Systems Biology, ChEM-H, and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA, USA
| | - Dinah M. Abeja
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Meredith N. Nix
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Jianing Zhong
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Mikołaj Słabicki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Eric S. Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Benjamin L. Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Nathanael S. Gray
- Department of Chemical and Systems Biology, ChEM-H, and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
29
|
Fan AT, Gadbois GE, Huang HT, Jiang J, Sigua LH, Smith ER, Wu S, Dunne-Dombrink K, Goyal P, Tao AJ, Sellers W, Fischer ES, Donovan KA, Ferguson FM. A Kinetic Scout Approach Accelerates Targeted Protein Degrader Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.612508. [PMID: 39345570 PMCID: PMC11429919 DOI: 10.1101/2024.09.17.612508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Bifunctional molecules such as targeted protein degraders induce proximity to promote gain-of-function pharmacology. These powerful approaches have gained broad traction across academia and the pharmaceutical industry, leading to an intensive focus on strategies that can accelerate their identification and optimization. We and others have previously used chemical proteomics to map degradable target space, and these datasets have been used to develop and train multiparameter models to extend degradability predictions across the proteome. In this study, we now turn our attention to develop generalizable chemistry strategies to accelerate the development of new bifunctional degraders. We implement lysine-targeted reversible-covalent chemistry to rationally tune the binding kinetics at the protein-of-interest across a set of 25 targets. We define an unbiased workflow consisting of global proteomics analysis, IP/MS of ternary complexes and the E-STUB assay, to mechanistically characterize the effects of ligand residence time on targeted protein degradation and formulate hypotheses about the rate-limiting step of degradation for each target. Our key finding is that target residence time is a major determinant of degrader activity, and this can be rapidly and rationally tuned through the synthesis of a minimal number of analogues to accelerate early degrader discovery and optimization efforts.
Collapse
Affiliation(s)
- Angela T. Fan
- Department of Chemistry and Biochemistry, University of California, San Diego
| | - Gillian E. Gadbois
- Department of Chemistry and Biochemistry, University of California, San Diego
| | | | - Jiewei Jiang
- Department of Chemistry and Biochemistry, University of California, San Diego
| | - Logan H. Sigua
- Medical Scientist Training Program, University of California, San Diego
| | - Emily R. Smith
- Department of Chemistry and Biochemistry, University of California, San Diego
| | - Sitong Wu
- Department of Chemistry and Biochemistry, University of California, San Diego
| | - Kara Dunne-Dombrink
- Department of Chemistry and Biochemistry, University of California, San Diego
| | - Pavitra Goyal
- Department of Chemistry and Biochemistry, University of California, San Diego
| | - Andrew J. Tao
- Department of Chemistry and Biochemistry, University of California, San Diego
| | | | - Eric S. Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston
| | - Katherine A. Donovan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston
| | - Fleur M. Ferguson
- Department of Chemistry and Biochemistry, University of California, San Diego
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego
| |
Collapse
|
30
|
Whittaker MK, Bendzunas GN, Shirani M, LeClair TJ, Shebl B, Dill TC, Coffino P, Simon SM, Kennedy EJ. Targeted Degradation of Protein Kinase A via a Stapled Peptide PROTAC. ACS Chem Biol 2024; 19:1888-1895. [PMID: 39137166 PMCID: PMC11420944 DOI: 10.1021/acschembio.4c00237] [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] [Indexed: 08/15/2024]
Abstract
Proteolysis-targeting chimeras (PROTACs) are bifunctional molecules that bind and recruit an E3 ubiquitin ligase to a targeted protein of interest, often through the utilization of a small molecule inhibitor. To expand the possible range of kinase targets that can be degraded by PROTACs, we sought to develop a PROTAC utilizing a hydrocarbon-stapled peptide as the targeting agent to bind the surface of a target protein of interest. In this study, we describe the development of a proteolysis-targeting chimera, dubbed Stapled Inhibitor Peptide - PROTAC or StIP-TAC, linking a hydrocarbon-stapled peptide with an E3 ligase ligand for targeted degradation of Protein Kinase A (PKA). This StIP-TAC molecule stimulated E3-mediated protein degradation of PKA, and this effect could be reversed by the addition of the proteasomal inhibitor MG-132. Further, StIP-TAC treatment led to a significant reduction in PKA substrate phosphorylation. Since many protein targets of interest lack structural features that make them amenable to small molecule targeting, development of StIP-TACs may broaden the potential range of protein targets using a PROTAC-mediated proteasomal degradation approach.
Collapse
Affiliation(s)
- Matthew K Whittaker
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| | - George N Bendzunas
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| | - Mahsa Shirani
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, New York 10065, United States
| | - Timothy J LeClair
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| | - Bassem Shebl
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, New York 10065, United States
| | - Taylor C Dill
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| | - Philip Coffino
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, New York 10065, United States
| | - Sanford M Simon
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, New York 10065, United States
| | - Eileen J Kennedy
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
31
|
Taylor JD, Barrett N, Martinez Cuesta S, Cassidy K, Pachl F, Dodgson J, Patel R, Eriksson TM, Riley A, Burrell M, Bauer C, Rees DG, Cimbro R, Zhang AX, Minter RR, Hunt J, Legg S. Targeted protein degradation using chimeric human E2 ubiquitin-conjugating enzymes. Commun Biol 2024; 7:1179. [PMID: 39300128 DOI: 10.1038/s42003-024-06803-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/29/2024] [Indexed: 09/22/2024] Open
Abstract
Proteins can be targeted for degradation by engineering biomolecules that direct them to the eukaryotic ubiquitination machinery. For instance, the fusion of an E3 ubiquitin ligase to a suitable target binding domain creates a 'biological Proteolysis-Targeting Chimera' (bioPROTAC). Here we employ an analogous approach where the target protein is recruited directly to a human E2 ubiquitin-conjugating enzyme via an attached target binding domain. Through rational design and screening we develop E2 bioPROTACs that induce the degradation of the human intracellular proteins SHP2 and KRAS. Using global proteomics, we characterise the target-specific and wider effects of E2 vs. VHL-based fusions. Taking SHP2 as a model target, we also employ a route to bioPROTAC discovery based on protein display libraries, yielding a degrader with comparatively weak affinity capable of suppressing SHP2-mediated signalling.
Collapse
Affiliation(s)
- Jonathan D Taylor
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK.
| | - Nathalie Barrett
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Sergio Martinez Cuesta
- Data Sciences and Quantitative Biology, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Katelyn Cassidy
- Protein Sciences, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Waltham, MA, 02451, USA
| | - Fiona Pachl
- Protein Sciences, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Waltham, MA, 02451, USA
| | - James Dodgson
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Radhika Patel
- Centre for Genomics Research, Dynamic Omics, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Tuula M Eriksson
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Aidan Riley
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Matthew Burrell
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Christin Bauer
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK
| | - D Gareth Rees
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Raffaello Cimbro
- Centre for Genomics Research, Dynamic Omics, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Andrew X Zhang
- Protein Sciences, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Waltham, MA, 02451, USA
| | - Ralph R Minter
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK
| | - James Hunt
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK.
| | - Sandrine Legg
- Biologics Engineering, R&D Oncology, AstraZeneca, Cambridge, CB2 0AA, UK
| |
Collapse
|
32
|
Gregory JA, Hickey CM, Chavez J, Cacace AM. New therapies on the horizon: Targeted protein degradation in neuroscience. Cell Chem Biol 2024; 31:1688-1698. [PMID: 39303702 DOI: 10.1016/j.chembiol.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/22/2024]
Abstract
This minireview explores the burgeoning field of targeted protein degradation (TPD) and its promising applications in neuroscience and clinical development. TPD offers innovative strategies for modulating protein levels, presenting a paradigm shift in small-molecule drug discovery and therapeutic interventions. Importantly, small-molecule protein degraders specifically target and remove pathogenic proteins from central nervous system cells without the drug delivery challenges of genomic and antibody-based modalities. Here, we review recent advancements in TPD technologies, highlight proteolysis targeting chimera (PROTAC) protein degrader molecules with proximity-induced degradation event-driven and iterative pharmacology, provide applications in neuroscience research, and discuss the high potential for translation of TPD into clinical settings.
Collapse
Affiliation(s)
| | | | - Juan Chavez
- Arvinas, Inc., 5 Science Park, New Haven, CT 06511, USA
| | | |
Collapse
|
33
|
Lampe GD, Liang AR, Zhang DJ, Fernández IS, Sternberg SH. Structure-guided engineering of type I-F CASTs for targeted gene insertion in human cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613948. [PMID: 39345383 PMCID: PMC11429998 DOI: 10.1101/2024.09.19.613948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Conventional genome editing tools rely on DNA double-strand breaks (DSBs) and host recombination proteins to achieve large insertions, resulting in a heterogeneous mixture of undesirable editing outcomes. We recently leveraged a type I-F CRISPR-associated transposase (CAST) from the Pseudoalteromonas Tn7016 transposon (PseCAST) for DSB-free, RNA-guided DNA integration in human cells, taking advantage of its programmability and large payload capacity. PseCAST is the only characterized CAST system that has achieved human genomic DNA insertions, but multiple lines of evidence suggest that DNA binding may be a critical bottleneck that limits high-efficiency activity. Here we report structural determinants of target DNA recognition by the PseCAST QCascade complex using single-particle cryogenic electron microscopy (cryoEM), which revealed novel subtype-specific interactions and RNA-DNA heteroduplex features. By combining our structural data with target DNA library screens and rationally engineered protein mutations, we uncovered CAST variants that exhibit increased integration efficiency and modified PAM stringency. Structure predictions of key interfaces in the transpososome holoenzyme also revealed opportunities for the design of hybrid CASTs, which we leveraged to build chimeric systems that combine high-activity DNA binding and DNA integration modules. Collectively, our work provides unique structural insights into type I-F CAST systems while showcasing multiple diverse strategies to investigate and engineer new RNA-guided transposase architectures for human genome editing applications.
Collapse
Affiliation(s)
- George D. Lampe
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Ashley R. Liang
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
- Present Address: Tornado Bio, San Francisco, CA, USA
| | - Dennis J. Zhang
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
- Present Address: Section of Microbiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Israel S. Fernández
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, Leioa, Spain
| | - Samuel H. Sternberg
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
- Howard Hughes Medical Institute, Columbia University, New York, NY, USA
| |
Collapse
|
34
|
Tsai JM, Nowak RP, Ebert BL, Fischer ES. Targeted protein degradation: from mechanisms to clinic. Nat Rev Mol Cell Biol 2024; 25:740-757. [PMID: 38684868 DOI: 10.1038/s41580-024-00729-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2024] [Indexed: 05/02/2024]
Abstract
Targeted protein degradation refers to the use of small molecules to induce the selective degradation of proteins. In its most common form, this degradation is achieved through ligand-mediated neo-interactions between ubiquitin E3 ligases - the principal waste disposal machines of a cell - and the protein targets of interest, resulting in ubiquitylation and subsequent proteasomal degradation. Notable advances have been made in biological and mechanistic understanding of serendipitously discovered degraders. This improved understanding and novel chemistry has not only provided clinical proof of concept for targeted protein degradation but has also led to rapid growth of the field, with dozens of investigational drugs in active clinical trials. Two distinct classes of protein degradation therapeutics are being widely explored: bifunctional PROTACs and molecular glue degraders, both of which have their unique advantages and challenges. Here, we review the current landscape of targeted protein degradation approaches and how they have parallels in biological processes. We also outline the ongoing clinical exploration of novel degraders and provide some perspectives on the directions the field might take.
Collapse
Affiliation(s)
- Jonathan M Tsai
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Radosław P Nowak
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Institute of Structural Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Boston, MA, USA.
| | - Eric S Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
35
|
Kadier K, Niu T, Ding B, Chen B, Qi X, Chen D, Cheng X, Fang Y, Zhou J, Zhao W, Liu Z, Yuan Y, Zhou Z, Dong X, Yang B, He Q, Cao J, Jiang L, Zhu CL. PROTAC-Mediated HDAC7 Protein Degradation Unveils Its Deacetylase-Independent Proinflammatory Function in Macrophages. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309459. [PMID: 39049738 PMCID: PMC11423193 DOI: 10.1002/advs.202309459] [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: 12/05/2023] [Revised: 04/25/2024] [Indexed: 07/27/2024]
Abstract
Class IIa histone deacetylases (Class IIa HDACs) play critical roles in regulating essential cellular metabolism and inflammatory pathways. However, dissecting the specific roles of each class IIa HDAC isoform is hindered by the pan-inhibitory effect of current inhibitors and a lack of tools to probe their functions beyond epigenetic regulation. In this study, a novel PROTAC-based compound B4 is developed, which selectively targets and degrades HDAC7, resulting in the effective attenuation of a specific set of proinflammatory cytokines in both lipopolysaccharide (LPS)-stimulated macrophages and a mouse model. By employing B4 as a molecular probe, evidence is found for a previously explored role of HDAC7 that surpasses its deacetylase function, suggesting broader implications in inflammatory processes. Mechanistic investigations reveal the critical involvement of HDAC7 in the Toll-like receptor 4 (TLR4) signaling pathway by directly interacting with the TNF receptor-associated factor 6 and TGFβ-activated kinase 1 (TRAF6-TAK1) complex, thereby initiating the activation of the downstream mitogen-activated protein kinase/nuclear factor-κB (MAPK/NF-κB) signaling cascade and subsequent gene transcription. This study expands the insight into HDAC7's role within intricate inflammatory networks and highlights its therapeutic potential as a novel target for anti-inflammatory treatments.
Collapse
Affiliation(s)
- Kailibinuer Kadier
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Tian Niu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Baoli Ding
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Boya Chen
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Xuxin Qi
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Danni Chen
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Xirui Cheng
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Yizheng Fang
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Jiahao Zhou
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Wenyi Zhao
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
| | - Zeqi Liu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Yi Yuan
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Zhan Zhou
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
| | - Xiaowu Dong
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, 310058, P. R. China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Cancer Center, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Bo Yang
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, 310058, P. R. China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Cancer Center, Zhejiang University, Hangzhou, 310058, P. R. China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, 310058, P. R. China
- School of Medicine, Hangzhou City University, Hangzhou, 310015, P. R. China
| | - Qiaojun He
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, 310058, P. R. China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Cancer Center, Zhejiang University, Hangzhou, 310058, P. R. China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, 310058, P. R. China
- Center for Drug Safety Evaluation and Research of Zhejiang University, Hangzhou, 310058, P. R. China
| | - Ji Cao
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, 310058, P. R. China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Cancer Center, Zhejiang University, Hangzhou, 310058, P. R. China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, 310058, P. R. China
| | - Li Jiang
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
| | - Cheng-Liang Zhu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, 310058, P. R. China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310018, P. R. China
- Cancer Center, Zhejiang University, Hangzhou, 310058, P. R. China
- Center for Drug Safety Evaluation and Research of Zhejiang University, Hangzhou, 310058, P. R. China
| |
Collapse
|
36
|
Huang HT, Lumpkin RJ, Tsai RW, Su S, Zhao X, Xiong Y, Chen J, Mageed N, Donovan KA, Fischer ES, Sellers WR. Ubiquitin-specific proximity labeling for the identification of E3 ligase substrates. Nat Chem Biol 2024; 20:1227-1236. [PMID: 38514884 DOI: 10.1038/s41589-024-01590-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 02/26/2024] [Indexed: 03/23/2024]
Abstract
Protein ubiquitylation controls diverse processes within eukaryotic cells, including protein degradation, and is often dysregulated in disease. Moreover, small-molecule degraders that redirect ubiquitylation activities toward disease targets are an emerging and promising therapeutic class. Over 600 E3 ubiquitin ligases are expressed in humans, but their substrates remain largely elusive, necessitating the development of new methods for their discovery. Here we report the development of E3-substrate tagging by ubiquitin biotinylation (E-STUB), a ubiquitin-specific proximity labeling method that biotinylates ubiquitylated substrates in proximity to an E3 ligase of interest. E-STUB accurately identifies the direct ubiquitylated targets of protein degraders, including collateral targets and ubiquitylation events that do not lead to substrate degradation. It also detects known substrates of E3 ligase CRBN and VHL with high specificity. With the ability to elucidate proximal ubiquitylation events, E-STUB may facilitate the development of proximity-inducing therapeutics and act as a generalizable method for E3-substrate mapping.
Collapse
Affiliation(s)
- Hai-Tsang Huang
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Ryan J Lumpkin
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Ryan W Tsai
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Shuyao Su
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Xu Zhao
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Yuan Xiong
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - James Chen
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Nada Mageed
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Katherine A Donovan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Eric S Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - William R Sellers
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
37
|
Yang J, Wu Y, Zhu Q, Qu X, Ou H, Liu H, Wei Y, Ge D, Lu C, Jiang B, Song X. Discovery of a first-in-class protein degrader for the c-ros oncogene 1 (ROS1). Bioorg Chem 2024; 150:107590. [PMID: 38955003 DOI: 10.1016/j.bioorg.2024.107590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/16/2024] [Accepted: 06/22/2024] [Indexed: 07/04/2024]
Abstract
The c-ros oncogene 1 (ROS1), an oncogenic driver, is known to induce non-small cell lung cancer (NSCLC) when overactivated, particularly through the formation of fusion proteins. Traditional targeted therapies focus on inhibiting ROS1 activity with ROS 1 inhibitors to manage cancer progression. However, a new strategy involving the design of protein degraders offers a more potent approach by completely degrading ROS1 fusion oncoproteins, thereby effectively blocking their kinase activity and enhancing anti-tumour potential. Utilizing PROteolysis-TArgeting Chimera (PROTAC) technology and informed by molecular docking and rational design, we report the first ROS1-specific PROTAC, SIAIS039. This degrader effectively targets multiple ROS1 fusion oncoproteins (CD74-ROS1, SDC4-ROS1 and SLC34A2-ROS1) in engineered Ba/F3 cells and HCC78 cells, demonstrating anti-tumour effects against ROS1 fusion-driven cancer cells. It suppresses cell proliferation, induces cell cycle arrest, and apoptosis, and inhibits clonogenicity. The anti-tumour efficacy of SIAIS039 surpasses two approved drugs, crizotinib and entrectinib, and matches that of the top inhibitors, including lorlatinib and taletrectinib. Mechanistic studies confirm that the degradation induced by 039 requires the participation of ROS1 ligands and E3 ubiquitin ligases, and involves the proteasome and ubiquitination. In addition, 039 exhibited excellent oral bioavailability in a mouse xenograft model, highlighting its potential for clinical application. In conclusion, our study presents a promising and novel therapeutic strategy for ROS1 fusion-positive NSCLC by targeting ROS1 fusion oncoproteins for degradation, laying the foundation for the development of further PROTAC and offering hope for patients with ROS1 fusion-positive NSCLC.
Collapse
Affiliation(s)
- Jiawen Yang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; Shanghai Clinical Research and Trial Center, Shanghai 201210, China
| | - Yifan Wu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qiaoliang Zhu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaojuan Qu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Hongyue Ou
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Haixia Liu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yongqi Wei
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Di Ge
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chunlai Lu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Biao Jiang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; CAS Key Laboratory of Synthetic Chemistry of Natural Substances, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China.
| | - Xiaoling Song
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
38
|
Jarusiewicz JA, Yoshimura S, Actis M, Li Y, Fu X, Yang L, Narina S, Pruett-Miller SM, Zhou S, Wang X, High AA, Nishiguchi G, Yang JJ, Rankovic Z. Development of an Orally Bioavailable LCK PROTAC Degrader as a Potential Therapeutic Approach to T-Cell Acute Lymphoblastic Leukemia. J Med Chem 2024; 67:11868-11884. [PMID: 38973320 DOI: 10.1021/acs.jmedchem.4c00481] [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: 07/09/2024]
Abstract
Despite significant advances over recent years, the treatment of T cell acute lymphoblastic leukemia (T-ALL) remains challenging. We have recently shown that a subset of T-ALL cases exhibited constitutive activation of the lymphocyte-specific protein tyrosine kinase (LCK) and were consequently responsive to treatments with LCK inhibitors and degraders such as dasatinib and dasatinib-based PROTACs. Here we report the design, synthesis and in vitro/vivo evaluation of SJ45566, a potent and orally bioavailable LCK PROTAC.
Collapse
Affiliation(s)
- Jamie A Jarusiewicz
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Satoshi Yoshimura
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Marisa Actis
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Yong Li
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Xiang Fu
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Lei Yang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Shilpa Narina
- Department of Cell and Molecular Biology and Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology and Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Suiping Zhou
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Xusheng Wang
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Anthony A High
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Gisele Nishiguchi
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Jun J Yang
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Zoran Rankovic
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| |
Collapse
|
39
|
Xiao Y, Yuan Y, Liu Y, Lin Z, Zheng G, Zhou D, Lv D. Targeted Protein Degradation: Current and Emerging Approaches for E3 Ligase Deconvolution. J Med Chem 2024; 67:11580-11596. [PMID: 38981094 DOI: 10.1021/acs.jmedchem.4c00723] [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] [Indexed: 07/11/2024]
Abstract
Targeted protein degradation (TPD), including the use of proteolysis-targeting chimeras (PROTACs) and molecular glue degraders (MGDs) to degrade proteins, is an emerging strategy to develop novel therapies for cancer and beyond. PROTACs or MGDs function by inducing the proximity between an E3 ligase and a protein of interest (POI), leading to ubiquitination and consequent proteasomal degradation of the POI. Notably, one major issue in TPD is the lack of ligandable E3 ligases, as current studies predominantly use CUL4CRBN and CUL2VHL. The TPD community is seeking to expand the landscape of ligandable E3 ligases, but most discoveries rely on phenotypic screens or serendipity, necessitating systematic target deconvolution. Here, we examine and discuss both current and emerging E3 ligase deconvolution approaches for degraders discovered from phenotypic screens or monovalent glue chemistry campaigns, highlighting future prospects for identifying more ligandable E3 ligases.
Collapse
Affiliation(s)
- Yufeng Xiao
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, 1333 Center Drive, Gainesville, Florida 32610, United States
| | - Yaxia Yuan
- Department of Biochemistry and Structural Biology and Center for Innovative Drug Discovery, School of Medicine, University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas 78229, United States
- Mays Cancer Center, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas 78229, United States
| | - Yi Liu
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, 1333 Center Drive, Gainesville, Florida 32610, United States
| | - Zongtao Lin
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, St. Louis, Missouri 63110, United States
| | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, 1333 Center Drive, Gainesville, Florida 32610, United States
| | - Daohong Zhou
- Department of Biochemistry and Structural Biology and Center for Innovative Drug Discovery, School of Medicine, University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas 78229, United States
- Mays Cancer Center, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas 78229, United States
| | - Dongwen Lv
- Department of Biochemistry and Structural Biology and Center for Innovative Drug Discovery, School of Medicine, University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas 78229, United States
- Mays Cancer Center, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas 78229, United States
| |
Collapse
|
40
|
Lynch TL, Marin VL, McClure RA, Phipps C, Ronau JA, Rouhimoghadam M, Adams AM, Kandi S, Wolke ML, Shergalis AG, Potts GK, Nacham O, Richardson P, Kakavas SJ, Chhor G, Jenkins GJ, Woller KR, Warder SE, Vasudevan A, Reitsma JM. Quantitative Measurement of Rate of Targeted Protein Degradation. ACS Chem Biol 2024; 19:1604-1615. [PMID: 38980123 DOI: 10.1021/acschembio.4c00262] [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: 07/10/2024]
Abstract
Targeted protein degradation (TPD) is a therapeutic approach that leverages the cell's natural machinery to degrade targets instead of inhibiting them. This is accomplished by using mono- or bifunctional small molecules designed to induce the proximity of target proteins and E3 ubiquitin ligases, leading to ubiquitination and subsequent proteasome-dependent degradation of the target. One of the most significant attributes of the TPD approach is its proposed catalytic mechanism of action, which permits substoichiometric exposure to achieve the desired pharmacological effects. However, apart from one in vitro study, studies supporting the catalytic mechanism of degraders are largely inferred based on potency. A more comprehensive understanding of the degrader catalytic mechanism of action can help aspects of compound development. To address this knowledge gap, we developed a workflow for the quantitative measurement of the catalytic rate of degraders in cells. Comparing a selective and promiscuous BTK degrader, we demonstrate that both compounds function as efficient catalysts of BTK degradation, with the promiscuous degrader exhibiting faster rates due to its ability to induce more favorable ternary complexes. By leveraging computational modeling, we show that the catalytic rate is highly dynamic as the target is depleted from cells. Further investigation of the promiscuous kinase degrader revealed that the catalytic rate is a better predictor of optimal degrader activity toward a specific target compared to degradation magnitude alone. In summary, we present a versatile method for mapping the catalytic activity of any degrader for TPD in cells.
Collapse
Affiliation(s)
- Thomas L Lynch
- Quantitative, Translational & ADME Sciences, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Violeta L Marin
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Ryan A McClure
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Colin Phipps
- Quantitative, Translational & ADME Sciences, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Judith A Ronau
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Milad Rouhimoghadam
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Ashley M Adams
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Soumya Kandi
- Quantitative, Translational & ADME Sciences, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Malerie L Wolke
- Quantitative, Translational & ADME Sciences, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Andrea G Shergalis
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Gregory K Potts
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Omprakash Nacham
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Paul Richardson
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Stephan J Kakavas
- Target Enabling Technologies, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Gekleng Chhor
- Target Enabling Technologies, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Gary J Jenkins
- Quantitative, Translational & ADME Sciences, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Kevin R Woller
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Scott E Warder
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Anil Vasudevan
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Justin M Reitsma
- Technology & Therapeutic Platforms, AbbVie Incorporated, 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| |
Collapse
|
41
|
HAMILTON GERHARD, EGGERSTORFER MARIETHERESE, STICKLER SANDRA. Development of PROTACS degrading KRAS and SOS1. Oncol Res 2024; 32:1257-1264. [PMID: 39055890 PMCID: PMC11267056 DOI: 10.32604/or.2024.051653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/24/2024] [Indexed: 07/28/2024] Open
Abstract
The Kirsten rat sarcoma virus-son of sevenless 1 (KRAS-SOS1) axis drives tumor growth preferentially in pancreatic, colon, and lung cancer. Now, KRAS G12C mutated tumors can be successfully treated with inhibitors that covalently block the cysteine of the switch II binding pocket of KRAS. However, the range of other KRAS mutations is not amenable to treatment and the G12C-directed agents Sotorasib and Adragrasib show a response rate of only approximately 40%, lasting for a mean period of 8 months. One approach to increase the efficacy of inhibitors is their inclusion into proteolysis-targeting chimeras (PROTACs), which degrade the proteins of interest and exhibit much higher antitumor activity through multiple cycles of activity. Accordingly, PROTACs have been developed based on KRAS- or SOS1-directed inhibitors coupled to either von Hippel-Lindau (VHL) or Cereblon (CRBN) ligands that invoke the proteasomal degradation. Several of these PROTACs show increased activity in vitro and in vivo compared to their cognate inhibitors but their toxicity in normal tissues is not clear. The CRBN PROTACs containing thalidomide derivatives cannot be tested in experimental animals. Resistance to such PROTACS arises through downregulation or inactivation of CRBN or factors of the functional VHL E3 ubiquitin ligase. Although highly active KRAS and SOS1 PROTACs have been formulated their clinical application remains difficult.
Collapse
Affiliation(s)
- GERHARD HAMILTON
- Institute of Pharmacology, Medical University of Vienna, Vienna, 1090, Austria
| | | | - SANDRA STICKLER
- Institute of Pharmacology, Medical University of Vienna, Vienna, 1090, Austria
| |
Collapse
|
42
|
Sobierajski T, Małolepsza J, Pichlak M, Gendaszewska-Darmach E, Błażewska KM. The impact of E3 ligase choice on PROTAC effectiveness in protein kinase degradation. Drug Discov Today 2024; 29:104032. [PMID: 38789027 DOI: 10.1016/j.drudis.2024.104032] [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: 02/02/2024] [Revised: 04/30/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024]
Abstract
Proteolysis targeting chimera (PROTACs) provide a novel therapeutic approach that is revolutionizing drug discovery. The success of PROTACs largely depends on the combination of their three fragments: E3 ligase ligand, linker and protein of interest (POI)-targeting ligand. We summarize the pivotal significance of the precise combination of the E3 ligase ligand with the POI-recruiting warhead, which is crucial for the successful execution of cellular processes and achieving the desired outcomes. Therefore, the key to our selection was the use of at least two ligands recruiting two different ligases. This approach enables a direct comparison of the impacts of the specific ligases on target degradation.
Collapse
Affiliation(s)
- Tomasz Sobierajski
- Institute of Organic Chemistry, Lodz University of Technology, Łódź, Poland
| | - Joanna Małolepsza
- Institute of Organic Chemistry, Lodz University of Technology, Łódź, Poland
| | - Marta Pichlak
- Institute of Molecular and Industrial Biotechnology, Lodz University of Technology, Łódź, Poland
| | | | | |
Collapse
|
43
|
Koldenhof P, Bemelmans MP, Ghosh B, Damm-Ganamet KL, van Vlijmen HWT, Pande V. Application of AlphaFold models in evaluating ligandable cysteines across E3 ligases. Proteins 2024; 92:819-829. [PMID: 38337153 DOI: 10.1002/prot.26675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/12/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024]
Abstract
Proteolysis Targeting Chimeras (PROTACs) are an emerging therapeutic modality and chemical biology tools for Targeted Protein Degradation (TPD). PROTACs contain a ligand targeting the protein of interest, a ligand recruiting an E3 ligase and a linker connecting these two ligands. There are over 600 E3 ligases known so far, but only a handful have been exploited for TPD applications. A key reason for this is the scarcity of ligands binding various E3 ligases and the paucity of structural data available, which complicates ligand design across the family. In this study, we aim to progress PROTAC discovery by proposing a shortlist of E3 ligases that can be prioritized for covalent targeting by performing systematic structural ligandability analysis on a chemoproteomic dataset of potentially reactive cysteines across hundreds of E3 ligases. One of the goals of this study is to apply AlphaFold (AF) models for ligandability evaluations, as for a vast majority of these ligases an experimental structure is not available in the protein data bank (PDB). Using a combination of pocket features, AF model quality and additional aspects, we propose a shortlist of E3 ligases and corresponding cysteines that can be prioritized to potentially discover covalent ligands and expand the PROTAC toolbox.
Collapse
Affiliation(s)
- Patrick Koldenhof
- Computer-Aided Drug Design, Janssen Pharmaceuticals, Beerse, Belgium
| | | | - Brahma Ghosh
- Discovery Chemistry, Janssen Pharmaceuticals, Spring House, Pennsylvania, USA
| | | | | | - Vineet Pande
- Computer-Aided Drug Design, Janssen Pharmaceuticals, Beerse, Belgium
| |
Collapse
|
44
|
Verma SK, Witkin KL, Sharman A, Smith MA. Targeting fusion oncoproteins in childhood cancers: challenges and future opportunities for developing therapeutics. J Natl Cancer Inst 2024; 116:1012-1018. [PMID: 38574391 PMCID: PMC11223828 DOI: 10.1093/jnci/djae075] [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: 02/01/2024] [Revised: 03/19/2024] [Accepted: 03/24/2024] [Indexed: 04/06/2024] Open
Abstract
Fusion oncoproteins are associated with childhood cancers and have proven challenging to target, aside from those that include kinases. As part of its efforts for targeting childhood cancers, the National Cancer Institute recently conducted a series on Novel Chemical Approaches for Targeting Fusion Oncoproteins. Key learnings on leading platforms and technologies that can be used to advance the development of molecular therapeutics that target fusion oncoproteins in childhood cancers are described. Recent breakthroughs in medicinal chemistry and chemical biology provide new ground and creative strategies to exploit for the development of targeted agents for improving outcomes against these recalcitrant cancers.
Collapse
Affiliation(s)
- Sharad K Verma
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Keren L Witkin
- Division of Cancer Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anu Sharman
- Division of Cancer Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Malcolm A Smith
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
45
|
Ichikawa S, Payne NC, Xu W, Chang CF, Vallavoju N, Frome S, Flaxman HA, Mazitschek R, Woo CM. The cyclimids: Degron-inspired cereblon binders for targeted protein degradation. Cell Chem Biol 2024; 31:1162-1175.e10. [PMID: 38320555 DOI: 10.1016/j.chembiol.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 11/02/2023] [Accepted: 01/11/2024] [Indexed: 02/08/2024]
Abstract
Cereblon (CRBN) is an E3 ligase substrate adapter widely exploited for targeted protein degradation (TPD) strategies. However, achieving efficient and selective target degradation is a preeminent challenge with ligands that engage CRBN. Here, we report that the cyclimids, ligands derived from the C-terminal cyclic imide degrons of CRBN, exhibit distinct modes of interaction with CRBN and offer a facile approach for developing potent and selective bifunctional degraders. Quantitative TR-FRET-based characterization of 60 cyclimid degraders in binary and ternary complexes across different substrates revealed that ternary complex binding affinities correlated strongly with cellular degradation efficiency. Our studies establish the unique properties of the cyclimids as versatile warheads in TPD and a systematic biochemical approach for quantifying ternary complex formation to predict their cellular degradation activity, which together will accelerate the development of ligands that engage CRBN.
Collapse
Affiliation(s)
- Saki Ichikawa
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - N Connor Payne
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA; Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Wenqing Xu
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Chia-Fu Chang
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Nandini Vallavoju
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Spencer Frome
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Hope A Flaxman
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Ralph Mazitschek
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Christina M Woo
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
46
|
Li Z, Liu HY, He Z, Chakravarty A, Golden RP, Jiang Z, You I, Yue H, Donovan KA, Du G, Che J, Tse J, Che I, Lu W, Fischer ES, Zhang T, Gray NS, Yang PL. Discovery of Potent Degraders of the Dengue Virus Envelope Protein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.01.596987. [PMID: 38854003 PMCID: PMC11160776 DOI: 10.1101/2024.06.01.596987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Targeted protein degradation has been widely adopted as a new approach to eliminate both established and previously recalcitrant therapeutic targets. Here we report the development of small molecule degraders of the envelope (E) protein of dengue virus. We developed two classes of bivalent E-degraders, linking two previously reported E-binding small molecules, GNF-2 and CVM-2-12-2, to a glutarimide-based recruiter of the CRL4CRBN ligase to effect proteosome-mediated degradation of the E protein. ZXH-2-107 (based on GNF-2) is an E degrader with ABL inhibition while ZXH-8-004 (based on CVM-2-12-2) is a selective and potent E-degrader. These two compounds provide proof-of-concept that difficult-to-drug targets such as a viral envelope protein can be effectively eliminated using a bivalent degrader and provide starting points for the future development of a new class antiviral drugs.
Collapse
Affiliation(s)
- Zhengnian Li
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Han-Yuan Liu
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA
| | - Zhixiang He
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA
| | - Antara Chakravarty
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA
| | - Ryan P. Golden
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Zixuan Jiang
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Inchul You
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Hong Yue
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Katherine A. Donovan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Guangyan Du
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Jianwei Che
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA
| | - Jason Tse
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Isaac Che
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Wenchao Lu
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Eric S. Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Tinghu Zhang
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Nathanael S. Gray
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Priscilla L. Yang
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
47
|
Reinecke M, Brear P, Vornholz L, Berger BT, Seefried F, Wilhelm S, Samaras P, Gyenis L, Litchfield DW, Médard G, Müller S, Ruland J, Hyvönen M, Wilhelm M, Kuster B. Chemical proteomics reveals the target landscape of 1,000 kinase inhibitors. Nat Chem Biol 2024; 20:577-585. [PMID: 37904048 PMCID: PMC11062922 DOI: 10.1038/s41589-023-01459-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/22/2023] [Indexed: 11/01/2023]
Abstract
Medicinal chemistry has discovered thousands of potent protein and lipid kinase inhibitors. These may be developed into therapeutic drugs or chemical probes to study kinase biology. Because of polypharmacology, a large part of the human kinome currently lacks selective chemical probes. To discover such probes, we profiled 1,183 compounds from drug discovery projects in lysates of cancer cell lines using Kinobeads. The resulting 500,000 compound-target interactions are available in ProteomicsDB and we exemplify how this molecular resource may be used. For instance, the data revealed several hundred reasonably selective compounds for 72 kinases. Cellular assays validated GSK986310C as a candidate SYK (spleen tyrosine kinase) probe and X-ray crystallography uncovered the structural basis for the observed selectivity of the CK2 inhibitor GW869516X. Compounds targeting PKN3 were discovered and phosphoproteomics identified substrates that indicate target engagement in cells. We anticipate that this molecular resource will aid research in drug discovery and chemical biology.
Collapse
Affiliation(s)
- Maria Reinecke
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
- German Cancer Consortium (DKTK), partner site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Paul Brear
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Larsen Vornholz
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), Munich, Germany
| | - Benedict-Tilmann Berger
- Structural Genomics Consortium, Buchmann Institute for Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Frankfurt, Germany
| | - Florian Seefried
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Stephanie Wilhelm
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Patroklos Samaras
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Laszlo Gyenis
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - David William Litchfield
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Guillaume Médard
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Susanne Müller
- Structural Genomics Consortium, Buchmann Institute for Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Frankfurt, Germany
| | - Jürgen Ruland
- German Cancer Consortium (DKTK), partner site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Marko Hyvönen
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Mathias Wilhelm
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
- Computational Mass Spectrometry, Technical University of Munich, Freising, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany.
- German Cancer Consortium (DKTK), partner site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Bavarian Biomolecular Mass Spectrometry Center (BayBioMS), Technical University of Munich, Freising, Germany.
| |
Collapse
|
48
|
Zhu D, Liang H, Du Z, Liu Q, Li G, Zhang W, Wu D, Zhou X, Song Y, Yang C. Altered Metabolism and Inflammation Driven by Post-translational Modifications in Intervertebral Disc Degeneration. RESEARCH (WASHINGTON, D.C.) 2024; 7:0350. [PMID: 38585329 PMCID: PMC10997488 DOI: 10.34133/research.0350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/18/2024] [Indexed: 04/09/2024]
Abstract
Intervertebral disc degeneration (IVDD) is a prevalent cause of low back pain and a leading contributor to disability. IVDD progression involves pathological shifts marked by low-grade inflammation, extracellular matrix remodeling, and metabolic disruptions characterized by heightened glycolytic pathways, mitochondrial dysfunction, and cellular senescence. Extensive posttranslational modifications of proteins within nucleus pulposus cells and chondrocytes play crucial roles in reshaping the intervertebral disc phenotype and orchestrating metabolism and inflammation in diverse contexts. This review focuses on the pivotal roles of phosphorylation, ubiquitination, acetylation, glycosylation, methylation, and lactylation in IVDD pathogenesis. It integrates the latest insights into various posttranslational modification-mediated metabolic and inflammatory signaling networks, laying the groundwork for targeted proteomics and metabolomics for IVDD treatment. The discussion also highlights unexplored territories, emphasizing the need for future research, particularly in understanding the role of lactylation in intervertebral disc health, an area currently shrouded in mystery.
Collapse
Affiliation(s)
- Dingchao Zhu
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Huaizhen Liang
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Zhi Du
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Qian Liu
- College of Life Sciences,
Wuhan University, Wuhan 430072, Hubei Province, China
| | - Gaocai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Weifeng Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Di Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Xingyu Zhou
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Yu Song
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| |
Collapse
|
49
|
Hernández-Morán BA, Taylor G, Lorente-Macías Á, Wood AJ. Degron tagging for rapid protein degradation in mice. Dis Model Mech 2024; 17:dmm050613. [PMID: 38666498 PMCID: PMC11073515 DOI: 10.1242/dmm.050613] [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] [Indexed: 05/01/2024] Open
Abstract
Degron tagging allows proteins of interest to be rapidly degraded, in a reversible and tuneable manner, in response to a chemical stimulus. This provides numerous opportunities for understanding disease mechanisms, modelling therapeutic interventions and constructing synthetic gene networks. In recent years, many laboratories have applied degron tagging successfully in cultured mammalian cells, spurred by rapid advances in the fields of genome editing and targeted protein degradation. In this At a Glance article, we focus on recent efforts to apply degron tagging in mouse models, discussing the distinct set of challenges and opportunities posed by the in vivo environment.
Collapse
Affiliation(s)
- Brianda A. Hernández-Morán
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road, Edinburgh EH4, 2XR, UK
| | - Gillian Taylor
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road, Edinburgh EH4, 2XR, UK
| | - Álvaro Lorente-Macías
- Edinburgh Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road, Edinburgh EH4 2XR, UK
| | - Andrew J. Wood
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road, Edinburgh EH4, 2XR, UK
| |
Collapse
|
50
|
Poirson J, Cho H, Dhillon A, Haider S, Imrit AZ, Lam MHY, Alerasool N, Lacoste J, Mizan L, Wong C, Gingras AC, Schramek D, Taipale M. Proteome-scale discovery of protein degradation and stabilization effectors. Nature 2024; 628:878-886. [PMID: 38509365 DOI: 10.1038/s41586-024-07224-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/22/2024] [Indexed: 03/22/2024]
Abstract
Targeted protein degradation and stabilization are promising therapeutic modalities because of their potency, versatility and their potential to expand the druggable target space1,2. However, only a few of the hundreds of E3 ligases and deubiquitinases in the human proteome have been harnessed for this purpose, which substantially limits the potential of the approach. Moreover, there may be other protein classes that could be exploited for protein stabilization or degradation3-5, but there are currently no methods that can identify such effector proteins in a scalable and unbiased manner. Here we established a synthetic proteome-scale platform to functionally identify human proteins that can promote the degradation or stabilization of a target protein in a proximity-dependent manner. Our results reveal that the human proteome contains a large cache of effectors of protein stability. The approach further enabled us to comprehensively compare the activities of human E3 ligases and deubiquitinases, identify and characterize non-canonical protein degraders and stabilizers and establish that effectors have vastly different activities against diverse targets. Notably, the top degraders were more potent against multiple therapeutically relevant targets than the currently used E3 ligases cereblon and VHL. Our study provides a functional catalogue of stability effectors for targeted protein degradation and stabilization and highlights the potential of induced proximity screens for the discovery of new proximity-dependent protein modulators.
Collapse
Affiliation(s)
- Juline Poirson
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Hanna Cho
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Akashdeep Dhillon
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Shahan Haider
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Ahmad Zoheyr Imrit
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Mandy Hiu Yi Lam
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Nader Alerasool
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Jessica Lacoste
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Lamisa Mizan
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Cassandra Wong
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Anne-Claude Gingras
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Daniel Schramek
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Mikko Taipale
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|