1
|
Qiu X, Zheng Q, Luo D, Ming Y, Zhang T, Pu W, Ai M, He J, Peng Y. Rational Design, Synthesis, and Biological Evaluation of Novel c-Met Degraders for Lung Cancer Therapy. J Med Chem 2025. [PMID: 39882685 DOI: 10.1021/acs.jmedchem.4c02129] [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
Cellular-mesenchymal epithelial transition factor (c-Met) is an attractive target for treating multiple cancers. Despite plentiful c-Met inhibitors have been developed, some issues, including the acquired drug resistance to c-Met inhibitors, have emerged to hamper their application in clinical treatment. Degradation of c-Met offers an opportunity to solve these issues. In this study, we developed a series of c-Met degraders, and the optimal compound 22b can efficiently degrade c-Met with a DC50 value of 0.59 nM in EBC-1 cells. Mechanistic studies revealed that compound 22b induced c-Met degradation via proteasome-mediated pathway. In addition, compound 22b suppressed the proliferation and also induced apoptosis of EBC-1 cells, outperforming the corresponding inhibitor tepotinib. Importantly, compound 22b showed favorable pharmacokinetic properties and significantly induced tumor regression in a xenograft model without obvious toxicity. In brief, this study provided compound 22b as a novel c-Met degrader for lung cancer therapy.
Collapse
Affiliation(s)
- Xingyang Qiu
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610064 Chengdu, China
| | - Qingquan Zheng
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610064 Chengdu, China
| | - Dongdong Luo
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610064 Chengdu, China
| | - Yue Ming
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610064 Chengdu, China
| | - Tingting Zhang
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610064 Chengdu, China
| | - Wenchen Pu
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610064 Chengdu, China
| | - Min Ai
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610064 Chengdu, China
| | - Jianhua He
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610064 Chengdu, China
| | - Yong Peng
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610064 Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, 610212 Chengdu, China
| |
Collapse
|
2
|
Gallo S, Folco CB, Crepaldi T. The MET Oncogene: An Update on Targeting Strategies. Pharmaceuticals (Basel) 2024; 17:1473. [PMID: 39598385 PMCID: PMC11597589 DOI: 10.3390/ph17111473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
The MET receptor, commonly known as HGF (hepatocyte growth factor) receptor, is a focus of extensive scientific research. MET has been linked to embryonic development, tissue regeneration following injury, tumorigenesis, and cancer metastasis. These functions underscore its involvement in numerous cellular processes, including stemness, proliferation, motility, cell dissociation, and survival. However, the enigmatic nature of MET becomes apparent in the context of cancer. When MET remains persistently activated, since its gene undergoes genetic alterations, it initiates a complex signaling cascade setting in motion an aggressive and metastatic program that is characteristic of malignant cells and is known as "invasive growth". The expanding knowledge of MET signaling has opened up numerous opportunities for therapeutic interventions, particularly in the realm of oncology. Targeting MET presents a promising strategy for developing novel anti-cancer treatments. In this review, we provide an updated overview of drugs designed to modulate MET signaling, highlighting MET kinase inhibitors, degraders, anti-MET/HGF monoclonal antibodies, and MET-targeted antibody-drug conjugates. Through this review, we aim to contribute to the ongoing advancement of therapeutic strategies targeting MET signaling.
Collapse
Affiliation(s)
- Simona Gallo
- Department of Oncology, University of Turin, Regione Gonzole 10, 10143 Orbassano, Italy; (S.G.); (C.B.F.)
- Candiolo Cancer Institute, FPO-IRCCS, SP142, Km 3.95, 10060 Candiolo, Italy
| | - Consolata Beatrice Folco
- Department of Oncology, University of Turin, Regione Gonzole 10, 10143 Orbassano, Italy; (S.G.); (C.B.F.)
- Candiolo Cancer Institute, FPO-IRCCS, SP142, Km 3.95, 10060 Candiolo, Italy
| | - Tiziana Crepaldi
- Department of Oncology, University of Turin, Regione Gonzole 10, 10143 Orbassano, Italy; (S.G.); (C.B.F.)
- Candiolo Cancer Institute, FPO-IRCCS, SP142, Km 3.95, 10060 Candiolo, Italy
| |
Collapse
|
3
|
Ying S, Chi H, Wu X, Zeng P, Chen J, Fu T, Fu W, Zhang P, Tan W. Selective and Orally Bioavailable c-Met PROTACs for the Treatment of c-Met-Addicted Cancer. J Med Chem 2024; 67:17053-17069. [PMID: 39348183 DOI: 10.1021/acs.jmedchem.3c02417] [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
c-Met is an attractive therapeutic target in multiple tumors. Previous studies have discovered some effective proteolysis-targeting chimeras (PROTACs) able to degrade c-Met; however, the structure-activity relationship (SAR), degradation selectivity, and pharmacokinetic profiles of c-Met PROTACs have, to date, remained largely unknown. Herein, through extensive SAR studies on various warheads, linkers, and E3 ligase ligands, a novel potent c-Met PROTAC Met-DD4 was identified. Our results suggested that Met-DD4 could induce robust c-Met degradation with excellent selectivity (DC50 = 6.21 nM), substantially killing the c-Met-addicted cancer cells (IC50 = 4.37 nM). Furthermore, in vivo studies showed that Met-DD4 could achieve excellent oral bioavailability and c-Met degradation, strongly retarding tumor growth with minute organ toxicity. Overall, this study reveals that targeted degradation of c-Met is a promising strategy for the treatment of c-Met-addicted cancers and provides novel lead compounds for the clinical translation of c-Met PROTACs.
Collapse
Affiliation(s)
- Shilong Ying
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Hongli Chi
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Xiaoqiu Wu
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Pingping Zeng
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Jinling Chen
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Ting Fu
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Weitao Fu
- Insitute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230601, China
| | - Penghui Zhang
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Weihong Tan
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
4
|
Stoikov II, Antipin IS, Burilov VA, Kurbangalieva AR, Rostovskii NV, Pankova AS, Balova IA, Remizov YO, Pevzner LM, Petrov ML, Vasilyev AV, Averin AD, Beletskaya IP, Nenajdenko VG, Beloglazkina EK, Gromov SP, Karlov SS, Magdesieva TV, Prishchenko AA, Popkov SV, Terent’ev AO, Tsaplin GV, Kustova TP, Kochetova LB, Magdalinova NA, Krasnokutskaya EA, Nyuchev AV, Kuznetsova YL, Fedorov AY, Egorova AY, Grinev VS, Sorokin VV, Ovchinnikov KL, Kofanov ER, Kolobov AV, Rusinov VL, Zyryanov GV, Nosov EV, Bakulev VA, Belskaya NP, Berezkina TV, Obydennov DL, Sosnovskikh VY, Bakhtin SG, Baranova OV, Doroshkevich VS, Raskildina GZ, Sultanova RM, Zlotskii SS, Dyachenko VD, Dyachenko IV, Fisyuk AS, Konshin VV, Dotsenko VV, Ivleva EA, Reznikov AN, Klimochkin YN, Aksenov DA, Aksenov NA, Aksenov AV, Burmistrov VV, Butov GM, Novakov IA, Shikhaliev KS, Stolpovskaya NV, Medvedev SM, Kandalintseva NV, Prosenko OI, Menshchikova EB, Golovanov AA, Khashirova SY. Organic Chemistry in Russian Universities. Achievements of Recent Years. RUSSIAN JOURNAL OF ORGANIC CHEMISTRY 2024; 60:1361-1584. [DOI: 10.1134/s1070428024080013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 03/28/2024] [Accepted: 03/31/2024] [Indexed: 01/06/2025]
|
5
|
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
|
6
|
Wang Z, Che S, Yu Z. PROTAC: Novel degradable approach for different targets to treat breast cancer. Eur J Pharm Sci 2024; 198:106793. [PMID: 38740076 DOI: 10.1016/j.ejps.2024.106793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/22/2024] [Accepted: 05/05/2024] [Indexed: 05/16/2024]
Abstract
The revolutionary Proteolysis Targeting Chimera (PROTACs) have the exciting potential to reshape the pharmaceutical industry landscape by leveraging the ubiquitin-proteasome system for targeted protein degradation. Breast cancer, the most prevalent cancer in women, could be treated using PROTAC therapy. Although substantial work has been conducted, there is not yet a comprehensive overview or progress update on PROTAC therapy for breast cancer. Hence, in this article, we've compiled recent research progress focusing on different breast cancer target proteins, such as estrogen receptor (ER), BET, CDK, HER2, PARP, EZH2, etc. This resource aims to serve as a guide for future PROTAC-based breast cancer treatment design.
Collapse
Affiliation(s)
- Zhenjie Wang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Office of Drug Clinical Trials, The People's Hospital of Gaozhou, Maoming, 525200, PR China
| | - Siyao Che
- Hepatological Surgery Department, The People's Hospital of Gaozhou, Maoming, 525200, PR China.
| | - Zhiqiang Yu
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan 523018, PR China.
| |
Collapse
|
7
|
Zhang R, Xie S, Ran J, Li T. Restraining the power of Proteolysis Targeting Chimeras in the cage: A necessary and important refinement for therapeutic safety. J Cell Physiol 2024; 239:e31255. [PMID: 38501341 DOI: 10.1002/jcp.31255] [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: 01/22/2024] [Revised: 03/02/2024] [Accepted: 03/06/2024] [Indexed: 03/20/2024]
Abstract
Proteolysis Targeting Chimeras (PROTACs) represent a significant advancement in therapeutic drug development by leveraging the ubiquitin-proteasome system to enable targeted protein degradation, particularly impacting oncology. This review delves into the various types of PROTACs, such as peptide-based, nucleic acid-based, and small molecule PROTACs, each addressing distinct challenges in protein degradation. It also discusses innovative strategies like bridged PROTACs and conditional switch-activated PROTACs, offering precise targeting of previously "undruggable" proteins. The potential of PROTACs extends beyond oncology, with ongoing research and technological advancements needed to maximize their therapeutic potential. Future progress in this field relies on interdisciplinary collaboration and the integration of advanced computational tools to open new treatment avenues across various diseases.
Collapse
Affiliation(s)
- Renshuai Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Songbo Xie
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Jie Ran
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Te Li
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
8
|
Jabbarzadeh Kaboli P, Chen HF, Babaeizad A, Roustai Geraylow K, Yamaguchi H, Hung MC. Unlocking c-MET: A comprehensive journey into targeted therapies for breast cancer. Cancer Lett 2024; 588:216780. [PMID: 38462033 DOI: 10.1016/j.canlet.2024.216780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/18/2024] [Accepted: 02/29/2024] [Indexed: 03/12/2024]
Abstract
Breast cancer is the most common malignancy among women, posing a formidable health challenge worldwide. In this complex landscape, the c-MET (cellular-mesenchymal epithelial transition factor) receptor tyrosine kinase (RTK), also recognized as the hepatocyte growth factor (HGF) receptor (HGFR), emerges as a prominent protagonist, displaying overexpression in nearly 50% of breast cancer cases. Activation of c-MET by its ligand, HGF, secreted by neighboring mesenchymal cells, contributes to a cascade of tumorigenic processes, including cell proliferation, metastasis, angiogenesis, and immunosuppression. While c-MET inhibitors such as crizotinib, capmatinib, tepotinib and cabozantinib have garnered FDA approval for non-small cell lung cancer (NSCLC), their potential within breast cancer therapy is still undetermined. This comprehensive review embarks on a journey through structural biology, multifaceted functions, and intricate signaling pathways orchestrated by c-MET across cancer types. Furthermore, we highlight the pivotal role of c-MET-targeted therapies in breast cancer, offering a clinical perspective on this promising avenue of intervention. In this pursuit, we strive to unravel the potential of c-MET as a beacon of hope in the fight against breast cancer, unveiling new horizons for therapeutic innovation.
Collapse
Affiliation(s)
- Parham Jabbarzadeh Kaboli
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan
| | - Hsiao-Fan Chen
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan
| | - Ali Babaeizad
- Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Hirohito Yamaguchi
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan; Department of Biotechnology, Asia University, Taichung, 413, Taiwan.
| |
Collapse
|
9
|
Dhull A, Wei J, Pulukuri AJ, Rani A, Sharma R, Mesbahi N, Yoon H, Savoy EA, Xaivong Vi S, Goody KJ, Berkman CE, Wu BJ, Sharma A. PSMA-targeted dendrimer as an efficient anticancer drug delivery vehicle for prostate cancer. NANOSCALE 2024; 16:5634-5652. [PMID: 38440933 DOI: 10.1039/d3nr06520k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Prostate cancer (PCa) is the second leading cause of cancer-related deaths among men in the United States. Although early-stage treatments exhibit promising 5-year survival rates, the treatment options for advanced stage disease are constrained, with short survival benefits due to the challenges associated with effective and selective drug delivery to PCa cells. Even though targeting Prostate Specific Membrane Antigen (PSMA) has been extensively explored and is clinically employed for imaging and radio-ligand therapy, the clinical success of PSMA-based approaches for targeted delivery of chemotherapies remains elusive. In this study, we combine a generation 4 hydroxy polyamidoamine dendrimer (PD) with irreversible PSMA ligand (CTT1298) to develop a PSMA-targeted nanoplatform (PD-CTT1298) for selective intracellular delivery of potent chemotherapeutics to PCa. PD-CTT1298-Cy5 exhibits a PSMA IC50 in the nanomolar range and demonstrates selective uptake in PSMA (+) PCa cells via PSMA mediated internalization. When systemically administered in a prostate tumor xenograft mouse model, PD-CTT1298-Cy5 selectively targets PSMA (+) tumors with significantly less accumulation in PSMA (-) tumors or upon blocking of the PSMA receptors. Moreover, the dendrimer clears rapidly from the off-target organs limiting systemic side-effects. Further, the conjugation of an anti-cancer agent, cabozantinib to the PSMA-targeted dendrimer translates to a significantly enhanced anti-proliferative activity in vitro compared to the free drug. These findings highlight the potential of PD-CTT1298 nanoplatform as a versatile approach for selective delivery of high payloads of potent chemotherapeutics to PCa, where dose related systemic side-effects are a major concern.
Collapse
Affiliation(s)
- Anubhav Dhull
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Jing Wei
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, USA
| | - Anunay James Pulukuri
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Anu Rani
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Rishi Sharma
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Nooshin Mesbahi
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Hosog Yoon
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Emily A Savoy
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Sylvia Xaivong Vi
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Kenneth John Goody
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Clifford E Berkman
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| | - Boyang Jason Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, USA
| | - Anjali Sharma
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA, USA.
| |
Collapse
|
10
|
Han X, Sun Y. PROTACs: A novel strategy for cancer drug discovery and development. MedComm (Beijing) 2023; 4:e290. [PMID: 37261210 PMCID: PMC10227178 DOI: 10.1002/mco2.290] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 06/02/2023] Open
Abstract
Proteolysis targeting chimera (PROTAC) technology has become a powerful strategy in drug discovery, especially for undruggable targets/proteins. A typical PROTAC degrader consists of three components: a small molecule that binds to a target protein, an E3 ligase ligand (consisting of an E3 ligase and its small molecule recruiter), and a chemical linker that hooks first two components together. In the past 20 years, we have witnessed advancement of multiple PROTAC degraders into the clinical trials for anticancer therapies. However, one of the major challenges of PROTAC technology is that only very limited number of E3 ligase recruiters are currently available as E3 ligand for targeted protein degradation (TPD), although human genome encodes more than 600 E3 ligases. Thus, there is an urgent need to identify additional effective E3 ligase recruiters for TPD applications. In this review, we summarized the existing RING-type E3 ubiquitin ligase and their small molecule recruiters that act as effective E3 ligands of PROTAC degraders and their application in anticancer drug discovery. We believe that this review could serve as a reference in future development of efficient E3 ligands of PROTAC technology for cancer drug discovery and development.
Collapse
Affiliation(s)
- Xin Han
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionChina National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational MedicineZhejiang University School of MedicineHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERZhejiang ProvinceChina
- Key Laboratory of Molecular Biology in Medical SciencesZhejiang ProvinceChina
| | - Yi Sun
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionChina National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational MedicineZhejiang University School of MedicineHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERZhejiang ProvinceChina
- Key Laboratory of Molecular Biology in Medical SciencesZhejiang ProvinceChina
- Research Center for Life Science and Human HealthBinjiang Institute of Zhejiang UniversityHangzhouChina
| |
Collapse
|