1
|
Phillips MJ, Alese OB, Horvat NK, Greene E, Gbolahan OB, Coleman K, Doxie DB, Parihar V, Mahdi ZK, McCook-Veal A, Switchenko JM, Diab M, Herting CJ, Paulos CM, El-Rayes BF, Lesinski GB. XL888 and pembrolizumab modulate the immune landscape of colorectal tumors in a phase Ib/II clinical trial. Oncoimmunology 2025; 14:2475620. [PMID: 40079916 PMCID: PMC11913390 DOI: 10.1080/2162402x.2025.2475620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/17/2025] [Accepted: 03/02/2025] [Indexed: 03/15/2025] Open
Abstract
We conducted a phase Ib/II clinical trial to evaluate the safety, feasibility, and clinical activity of combining pembrolizumab (anti-PD-1) with XL888 (Hsp90 inhibitor) in patients with advanced colorectal cancer (CRC). We hypothesized that this regimen would modulate soluble and cellular immune mediators and enhance clinical outcomes. The trial employed a 3 + 3 open-label design, with an expansion cohort at the recommended phase II dose (RP2D) in treatment-refractory, mismatch repair-proficient CRC patients. Comprehensive analyses of plasma cytokines, peripheral blood mononuclear cells (PBMCs), and spatial immune cell patterns in liver biopsies were performed to identify unique immune signatures resulting from the combined therapy. The combination of pembrolizumab and XL888 proved to be safe and feasible, with a subset of patients achieving stable disease, although no objective responses were observed in this heavily pre-treated population. Correlative studies revealed immunomodulatory effects in tumors and circulation, including a reduction in IL6+ cells and macrophages (CD68+) within metastatic liver tissue, alterations in blood CD3+ cells, and upregulation of numerous inflammatory plasma cytokines. These findings suggest local and systemic immune activation by the combination of pembrolizumab and XL888. While clinical activity was modest in treatment-refractory CRC patients, there were notable effects on the tumor immune environment and systemic immune modulation.
Collapse
Affiliation(s)
- Maggie J. Phillips
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Olatunji B. Alese
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Natalie K. Horvat
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Emily Greene
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Olumide B. Gbolahan
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | | | - Deon B. Doxie
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Vaunita Parihar
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Zaid K. Mahdi
- Department of Pathology, Emory University, Atlanta, GA, USA
| | - Ashley McCook-Veal
- Department of Biostatistics & Bioinformatics, Emory University, Atlanta, GA, USA
| | | | - Maria Diab
- Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Cameron J. Herting
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Chrystal M. Paulos
- Department of Surgery, Emory University, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Bassel F. El-Rayes
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gregory B. Lesinski
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| |
Collapse
|
2
|
Wang H, Zhang W, Sun Y, Xu X, Chen X, Zhao K, Yang Z, Liu H. Nanotherapeutic strategies exploiting biological traits of cancer stem cells. Bioact Mater 2025; 50:61-94. [PMID: 40242505 PMCID: PMC12002948 DOI: 10.1016/j.bioactmat.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/08/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer stem cells (CSCs) represent a distinct subpopulation of cancer cells that orchestrate cancer initiation, progression, metastasis, and therapeutic resistance. Despite advances in conventional therapies, the persistence of CSCs remains a major obstacle to achieving cancer eradication. Nanomedicine-based approaches have emerged for precise CSC targeting and elimination, offering unique advantages in overcoming the limitations of traditional treatments. This review systematically analyzes recent developments in nanomedicine for CSC-targeted therapy, emphasizing innovative nanomaterial designs addressing CSC-specific challenges. We first provide a detailed examination of CSC biology, focusing on their surface markers, signaling networks, microenvironmental interactions, and metabolic signatures. On this basis, we critically evaluate cutting-edge nanomaterial engineering designed to exploit these CSC traits, including stimuli-responsive nanodrugs, nanocarriers for drug delivery, and multifunctional nanoplatforms capable of generating localized hyperthermia or reactive oxygen species. These sophisticated nanotherapeutic approaches enhance selectivity and efficacy in CSC elimination, potentially circumventing drug resistance and cancer recurrence. Finally, we present an in-depth analysis of current challenges in translating nanomedicine-based CSC-targeted therapies from bench to bedside, offering critical insights into future research directions and clinical implementation. This review aims to provide a comprehensive framework for understanding the intersection of nanomedicine and CSC biology, contributing to more effective cancer treatment modalities.
Collapse
Affiliation(s)
- Hongyu Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Wenjing Zhang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Yun Sun
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xican Xu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xiaoyang Chen
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Kexu Zhao
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Zhao Yang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Huiyu Liu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| |
Collapse
|
3
|
Li X, Liu M, Wang H, Mao J, Zhai X, He T, Ma J, Zhou Y, Lu W, Yu X, Li J, Zhu S. Design, synthesis and biological evaluation of WEE1 degraders via HSP90-mediated targeting chimeras for target therapy of acute myeloid leukemia. Eur J Med Chem 2025; 290:117512. [PMID: 40112662 DOI: 10.1016/j.ejmech.2025.117512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
Targeted protein degradation (TPD) technology is a promising strategy for drug development, while the on-target off-tumor risks of current TPD technologies were intractable. Herein, a series of (HSP90)-mediated targeting chimeras (HEMTACs) based WEE1-target degraders were designed to enhance the efficiency and decrease off-tumor risks. Among them, 8b and 9c could effectively degrade cellular WEE1 protein and exhibited superior anti-proliferative activity in MV-4-11 cells by inducing cell cycle arrest in G2/M phase. Meanwhile, 8b and 9c exhibited high selectivity to primary AML cells over normal cells. Furthermore, 3 mg/kg of 9c demonstrated superior anti-cancer activity than 5 mg/kg AZD1775 in an AML PDX model. And most importantly, 9c exhibited lower hematotoxicity than equimolar AZD1775 in mice safety evaluation, suggesting that 9c is a promising degrader for AML target therapy, comfirming that HSP90-based HEMTACs is a valid strategy to reduce off-tumor risks.
Collapse
Affiliation(s)
- Xiaomei Li
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, PR China
| | - Minmin Liu
- State Key Laboratory of Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Media, Chinese Academy of Sciences, Shanghai, 201203, PR China
| | - Huijing Wang
- School of Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Rd, Nanjing, 210023, PR China
| | - Jialuo Mao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Xiangying Zhai
- State Key Laboratory of Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Media, Chinese Academy of Sciences, Shanghai, 201203, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; Zhongshan Institute of Drug Discovery, Institution for Drug Discovery Innovation, Chinese Academy of Science, Zhongshan Tsuihang New District, Guangdong Province, 528400, PR China
| | - Tingting He
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, PR China
| | - Jingkun Ma
- State Key Laboratory of Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Media, Chinese Academy of Sciences, Shanghai, 201203, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Yubo Zhou
- State Key Laboratory of Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Media, Chinese Academy of Sciences, Shanghai, 201203, PR China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; Zhongshan Institute of Drug Discovery, Institution for Drug Discovery Innovation, Chinese Academy of Science, Zhongshan Tsuihang New District, Guangdong Province, 528400, PR China
| | - Wei Lu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, PR China
| | - Xiaoxuan Yu
- School of Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Rd, Nanjing, 210023, PR China; State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 163 Xianlin Road, Nanjing, 210023, PR China.
| | - Jia Li
- State Key Laboratory of Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Media, Chinese Academy of Sciences, Shanghai, 201203, PR China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China.
| | - Shulei Zhu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, PR China; Innovation Center for AI and Drug Discovery, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, PR China.
| |
Collapse
|
4
|
Feng ZW, Li L, Zhang SD, Wang YJ, Pei JY, Chen NN, Wu BD, Zheng QL, You QD, Guo XK, Xu XL. Structural Optimization of Pyrazole Compounds as Hsp90 Regulators with Enhanced Antitumor Activity. J Med Chem 2025; 68:9886-9905. [PMID: 40310691 DOI: 10.1021/acs.jmedchem.4c02182] [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: 05/03/2025]
Abstract
Targeting Hsp90 is an effective strategy for cancer therapy. TAS-116 has been approved for the treatment of gastrointestinal stromal tumors. Our previous studies identified a series of pyrazole derivatives as covalent Hsp90 inhibitors that allosterically disrupt the Hsp90-Cdc37 interaction. Here, through systematic structure-activity relationship (SAR) optimization, compound 39 (DDO-6691) with a new covalent warhead was developed, which demonstrates improved ADME properties and significantly enhanced antitumor activity. Notably, parental HCT-116 cells exhibited markedly greater sensitivity to compound 39 (IC50 > 50 μM) compared to their Cdc37-knockout counterparts. Importantly, compound 39 displayed potent tumor growth inhibition in HCT-116 xenograft mouse models. These collective findings underscore the therapeutic promise of covalent Hsp90-targeted disruption of the Hsp90-Cdc37 complex, offering a novel mechanistic approach to cancer treatment.
Collapse
Affiliation(s)
- Zi-Wen Feng
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Li Li
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- School of Pharmacy, Jiangsu Health Vocational College, Nanjing 210029, China
| | - Shi-Duo Zhang
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ying-Ji Wang
- Department of Pharmaceutical Analysis, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jia-Yue Pei
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Nan-Nan Chen
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Bei-Duo Wu
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qiu-Ling Zheng
- Department of Pharmaceutical Analysis, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qi-Dong You
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao-Ke Guo
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao-Li Xu
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
5
|
Kowalewski ME, Zagler S, Redinbo MR. Structural Insights into Selectively Targeting Candida albicans Hsp90. Biochemistry 2025. [PMID: 40397669 DOI: 10.1021/acs.biochem.5c00015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
The threat of drug-resistant pathogens continues to rise and underscores the need for new antimicrobial and antifungal strategies. Diverse chemical scaffolds have been shown with high affinity to bind the human heat-shock protein Hsp90. Orthologous proteins are present in microbial pathogens and have been shown to be particularly abundant in these organisms, suggesting they may serve as therapeutic targets. Here, we examine the potency and selectivity of human Hsp90 ligands for their capacity to bind to the nucleotide binding domain of Hsp90 from the pathogenic fungi, Candida albicans. Using a series of biochemical, structural, and fragment and in silico screening investigations, we define key chemical features that lead to effective C. albicans Hsp90 (CaHsp90) binding. We support these studies with crystal structures of five diverse human Hsp90 ligands in complex with CaHsp90, as well as the structure of this protein with a nonhydrolyzable ATP analog. We demonstrate the structural basis for the selectivity of the human Hsp90 inhibitor TAS116 for CaHsp90, features that may be exploited in the future development of improved CaHsp90 inhibitors.
Collapse
Affiliation(s)
- Mark E Kowalewski
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Sebastian Zagler
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Matthew R Redinbo
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina 27599, United States
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
- Department of Microbiology and Immunology, and Integrative Program for Biological and Genome Sciences, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
6
|
Chen X, Chen Z, Xu Y, Zou P, Shen W, Zhang Z, Wang Y. Genome-wide identification of heat shock protein 90 family in Larimichthys crocea and expression analysis in response to thermal stress and Vibrio parahaemolyticus infection. Comp Biochem Physiol B Biochem Mol Biol 2025; 279:111112. [PMID: 40398836 DOI: 10.1016/j.cbpb.2025.111112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 05/14/2025] [Accepted: 05/15/2025] [Indexed: 05/23/2025]
Abstract
Members of the heat shock protein 90 family (HSP90s) are evolutionarily conserved and play crucial roles in protein transport, immune regulation and antigen presentation. In this study, five hsp90s were identified from the genome of large yellow croaker (Larimichthys crocea) and analyzed using bioinformatics. All five identified hsp90s encode proteins with HATPase_c and HSP90 domains, and are mainly localized in the cytoplasm, mitochondria and endoplasmic reticulum. Chromosomal mapping revealed their distribution across three distinct chromosomes. Quantitative real-time PCR (qPCR) analysis showed differential expression patterns of the five hsp90s in 11 tissues. Additionally, their expression dynamics in the liver, spleen, head kidney, gill and blood were analyzed at 3 h, 12 h, 24 h and 48 h post thermal stress, Vibrio parahaemolyticus infection or under a combination of these two stressors. Results showed that the L. crocea hsp90s exhibited distinct expression patterns in response to the above three stimuli in different immune tissues. Notably, hsp90s in the spleen were most responsive. This study systematically clarified for the first time the gene structure characteristics, tissue expression patterns, and environmental stress response mechanisms of the HSP90 family in L. crocea. It confirmed that hsp90s show significant functional differentiation and synergy in response to biotic (pathogen infection) and abiotic (thermal stress) stresses, and provides important clues for a deeper understanding of the genetic basis of environmental adaptation in L. crocea.
Collapse
Affiliation(s)
- Xinxin Chen
- State Key Laboratory of Mariculture Breeding, Jimei University, Xiamen 361021, China; Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen 361021, China
| | - Zebin Chen
- State Key Laboratory of Mariculture Breeding, Jimei University, Xiamen 361021, China; Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen 361021, China
| | - Yuqing Xu
- State Key Laboratory of Mariculture Breeding, Jimei University, Xiamen 361021, China; Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen 361021, China
| | - Pengfei Zou
- State Key Laboratory of Mariculture Breeding, Jimei University, Xiamen 361021, China; Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen 361021, China
| | - Weiliang Shen
- Ningbo Academy of Oceanology and Fishery, Ningbo 315048, China
| | - Ziping Zhang
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Mariculture Breeding, Fujian Agriculture and Forestry University, Fuzhou, China, Fuzhou 350002, China.
| | - Yilei Wang
- State Key Laboratory of Mariculture Breeding, Jimei University, Xiamen 361021, China; Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen 361021, China.
| |
Collapse
|
7
|
Shahana MV, Choudhary B. HSP90 and the cancer transcriptome: a comprehensive review of inhibitors and mechanistic insights. Int J Clin Oncol 2025:10.1007/s10147-025-02782-6. [PMID: 40383747 DOI: 10.1007/s10147-025-02782-6] [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: 11/19/2024] [Accepted: 05/01/2025] [Indexed: 05/20/2025]
Abstract
This review summarizes the structure, function, expression, and inhibitors of HSP90, the chaperone, in cancers. It systematically investigates the effects of HSP90 inhibitors, including AUY922, B11B021, CCT-018159, D7-gedunin, geldanamycin, and gedunin, across a range of cancer cell lines (HCC151, HT29, MCF7, PC3, VCAP, and A375) and a normal HA1E cell line, using data from the CLUE database. Our analysis reveals that treatment with these HSP90 inhibitors induces significant stress responses in tumor cells, initiating intrinsic and extrinsic apoptotic pathways. The HSP90AA1, HSP90AB1, HSP27, HSP70, VEGF, and NOTCH exhibited notable upregulation at 24 h post-treatment compared to 6 h, indicating a time-dependent increase in cellular stress (heat shock response) and activation of pro-survival signaling mechanisms. Additionally, the study highlights a significant upregulation of immune-related pathways, including those involving IL10, IL3, and IL7, following HSP90 inhibition, indicating that these inhibitors not only directly affect tumor cell viability but also modulate the tumor microenvironment by enhancing immune cell activation and cytokine release. The elevated levels of IL10 point to a dual role, where immune suppression mechanisms are also at play, potentially facilitating immune evasion by the tumor. The findings suggest that HSP90 inhibitors exhibit varying mechanisms of action across different cancer cell lines despite the presence of some common targets. These insights highlight the need for further investigation into the precise mechanisms of HSP90 inhibitors to optimize their therapeutic potential in different cancers.
Collapse
Affiliation(s)
- M V Shahana
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore, Karnataka, 560100, India
- Research scholar, Manipal Academy of Higher Education (MAHE), Manipal, 576104, India
| | - Bibha Choudhary
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore, Karnataka, 560100, India.
| |
Collapse
|
8
|
Liu J, Su Y, Zhang C, Dong H, Yu R, Yang X, Tian Y, Feng Y, Zhang J, Shi M, Wang C, Li W, Liu J, He L, Yang X, Liu H. NCOA3 impairs the efficacy of anti-PD-L1 therapy via HSP90α/EZH2/CXCL9 axis in colon cancer. Int Immunopharmacol 2025; 155:114579. [PMID: 40215778 DOI: 10.1016/j.intimp.2025.114579] [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: 12/05/2024] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized colon cancer treatment, but their efficacy is largely restricted by the limited presence of CD8+ cytotoxic T lymphocytes (CTLs). However, the specific genetic alterations that impact the CD8+ CTL infiltration in colon cancer remain poorly understood. Here, we analyzed clinical and multi-omics data from the Memorial Sloan-Kettering Cancer Center (MSKCC) ICIs-treated and The Cancer Genome Atlas (TCGA) colon adenocarcinoma (COAD) cohorts to screen the key mutations that may influence the efficacy of immunotherapy. We found that patients with NCOA3 mutations exhibit better response to immunotherapy and higher CD8+ CTL infiltration. In vitro and in vivo experiments revealed that mutant NCOA3 increases the efficacy of anti-PD-L1 and CD8+ CTL recruitment by upregulating C-X-C motif chemokine ligand 9 (CXCL9), which is dependent on its impaired intrinsic histone acetyltransferase activity. Mechanistically, wild-type NCOA3 as histone acetyltransferase upregulates Heat shock protein 90 alpha (HSP90α) by enhancing histone H3 lysine 27 acetylation (H3K27ac) at its promoter region. Increased HSP90α stabilizes Enhancer of zeste homolog 2 (EZH2), which then increase the histone H3 lysine 27 trimethylation (H3K27me3) at the CXCL9 promoter region, thereby suppressing the expression of CXCL9. Targeted inhibition of NCOA3 by small molecular inhibitor SI-2 improves the efficacy of PD-L1 blockade therapy. NCOA3 could serve as a novel biomarker and potential target to improve the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Jiaqi Liu
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Yixi Su
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Chi Zhang
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Haiyan Dong
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Runfeng Yu
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xin Yang
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Yu Tian
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Yanchun Feng
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Jingdan Zhang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Mengchen Shi
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Chen Wang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Weiqian Li
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Jun Liu
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Lingyuan He
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xiangling Yang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.
| | - Huanliang Liu
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.
| |
Collapse
|
9
|
Chen J, Zhang H, Qiu M, Hu J, Lin L, Mai L, Huang G, Chen X, Li X, Qin X, Zhao H. Honokiol in the treatment of triple-negative breast cancer: a network pharmacology approach and experimental validation. Biochem Biophys Res Commun 2025; 771:152008. [PMID: 40398092 DOI: 10.1016/j.bbrc.2025.152008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 05/12/2025] [Accepted: 05/12/2025] [Indexed: 05/23/2025]
Abstract
Triple-negative breast cancer (TNBC) is a rare and highly metastatic form of cancer. Honokiol (HNK), a biphenolic compound, has been utilized in TNBC treatment, though its specific targets remain unclear. This study aimed to elucidate the effects of HNK on TNBC by combining network pharmacology predictions and experimental validation to uncover its mechanisms. MDA-MB 231 and MDA-MB 468 cells were pre-treated with varying doses of HNK for 24 h. Cell viability, proliferation, and apoptosis were assessed using CCK8 and FACS assays, whereas a wound healing assay was used to evaluate cell migration. A tubule formation assay was used to assess blood vessel formation in HUVECs. Additionally, in vivo activity was confirmed using a zebrafish xenograft model. Network pharmacology and molecular docking predicted active ingredients, key targets, and potential mechanisms of HNK against TNBC. Results indicated that HNK induces apoptosis in MDA-MB 231 and MDA-MB 468 cells and inhibits their migration and proliferation. Furthermore, HNK suppressed blood vessel formation. Zebrafish xenograft experiments validated HNK's inhibitory effect on TNBC cells in vivo. Network pharmacology identified 36 potential HNK targets against TNBC, including HSP90AA1, AKT1, EGFR, ERBB2, HSP90AB1, PGR, MDM2, HDAC1, NR3C1, and MAPK14. Key signaling pathways such as PI3K-Akt, MAPK, Rap1, Ras, and FoxO were implicated in HNK's anti-TNBC mechanism. Molecular docking demonstrated spontaneous interactions between HNK and the targeted proteins. In conclusion, HNK may reduce angiogenesis by blocking the EGFR and HSP90AB1 pathways thereby decreasing proliferation and increasing apoptosis in TNBC cells.
Collapse
Affiliation(s)
- Jing Chen
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China; Medical Research Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China
| | - Haipeng Zhang
- Department of Blood Transfusion, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China
| | - Min Qiu
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China
| | - Jiemei Hu
- Department of Gynecology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China
| | - Lu Lin
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China
| | - Liping Mai
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China
| | - Guiping Huang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China
| | - Xiuyun Chen
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China
| | - Xiaohong Li
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China
| | - Xianyu Qin
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, PR China.
| | - Haishan Zhao
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, PR China.
| |
Collapse
|
10
|
Pasala C, Digwal CS, Sharma S, Wang S, Bubula A, Chiosis G. Epichaperomes: redefining chaperone biology and therapeutic strategies in complex diseases. RSC Chem Biol 2025; 6:678-698. [PMID: 40144950 PMCID: PMC11933791 DOI: 10.1039/d5cb00010f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025] Open
Abstract
The complexity of disease biology extends beyond mutations or overexpression, encompassing stress-induced mechanisms that reshape proteins into pathological assemblies. Epichaperomes, stable and disease-specific assemblies of chaperones and co-chaperones, exemplify this phenomenon. This review emphasizes the critical structural and functional distinctions between epichaperomes and canonical chaperones, highlighting their role in redefining therapeutic strategies. Epichaperomes arise under stress conditions through post-translational modifications that stabilize these assemblies, enabling them to act as scaffolding platforms that rewire protein-protein interaction networks and drive the pathological phenotypes of complex diseases such as cancer and neurodegeneration. Chemical biology has been instrumental in uncovering the unique nature of epichaperomes, with small molecules like PU-H71 elucidating their biology and demonstrating their therapeutic potential by dismantling pathological scaffolds and restoring normal protein-protein interaction networks. By targeting epichaperomes, we unlock the potential for network-level interventions and personalized medicine, offering transformative possibilities for diseases driven by protein-protein interaction network dysregulation.
Collapse
Affiliation(s)
- Chiranjeevi Pasala
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center New York NY 10065 USA
| | - Chander S Digwal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center New York NY 10065 USA
| | - Sahil Sharma
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center New York NY 10065 USA
| | - Shujuan Wang
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center New York NY 10065 USA
| | - Alessia Bubula
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center New York NY 10065 USA
| | - Gabriela Chiosis
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center New York NY 10065 USA
- Department of Medicine, Division of Solid Tumors, Memorial Sloan Kettering Cancer Center New York NY 10065 USA
| |
Collapse
|
11
|
Peng B, Wang Y, Zhang H. Mitonuclear Communication in Stem Cell Function. Cell Prolif 2025; 58:e13796. [PMID: 39726221 DOI: 10.1111/cpr.13796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/25/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024] Open
Abstract
Mitochondria perform multiple functions within the cell, including the production of ATP and a great deal of metabolic intermediates, while also contributing to the cellular stress response. The majority of mitochondrial proteins are encoded by nuclear genomes, highlighting the importance of mitonuclear communication for sustaining mitochondrial homeostasis and functional. As a crucial part of the intracellular signalling network, mitochondria can impact stem cell fate determinations. Considering the essential function of stem cells in tissue maintenance, regeneration and aging, it is important to understand how mitochondria influence stem cell fate. This review explores the significant roles of mitonuclear communication and mitochondrial proteostasis, highlighting their influence on stem cells. We also examine how mitonuclear interactions contribute to cellular homeostasis, stem cell therapies, and the potential for extending lifespan.
Collapse
Affiliation(s)
- Baozhou Peng
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- The Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yaning Wang
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- The Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Hongbo Zhang
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- The Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
12
|
Chaudhury S, D'Amico T, Blagg BSJ. The Hsp90β Isoform: An Attractive Target for Drug Development. Med Res Rev 2025. [PMID: 40293270 DOI: 10.1002/med.22114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 02/12/2025] [Accepted: 04/09/2025] [Indexed: 04/30/2025]
Abstract
The beta isoform of 90 kDa heat shock protein (Hsp90β) plays a critical role in maintaining cellular proteostasis by assisting in the folding and refolding of proteins, which is essential for both normal cellular function and stress response. It is constitutively expressed in mammalian cells, differentiating it from the inducible Hsp90α isoform. Hsp90β's involvement in diverse cellular processes, such as signal transduction, cell cycle control, and apoptosis, underscores its significant role in various diseases, including cancer and neurodegenerative disorders. The isoform-specific functions of Hsp90β and its interaction with unique client proteins make it a promising target for therapeutic intervention, particularly in the development of selective inhibitors that avoid the adverse effects observed with pan-Hsp90 inhibitors. This review delves into the structural and functional intricacies of Hsp90β, its role in disease, and the potential for selective drug development.
Collapse
Affiliation(s)
- Subhabrata Chaudhury
- Department of Chemistry and Biochemistry, Warren Family Research Center for Drug Discovery and Development, University of Notre Dame, Notre Dame, Indiana, USA
| | - Terin D'Amico
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Brian S J Blagg
- Department of Chemistry and Biochemistry, Warren Family Research Center for Drug Discovery and Development, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
13
|
Ding M, Chen H, He L, Wang Z, Zhao X, Sun P, Mei Q, Li D, Fan Q. NIR-II D-A-D-Type Small-Molecule Coordination with Carboxylatopillar[5]Arene: a Multifunctional Phototheranostic for Low-Temperature NIR-II Photothermal/Platinum-Based/Chemodynamic Combination Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2501903. [PMID: 40255101 DOI: 10.1002/smll.202501903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/03/2025] [Indexed: 04/22/2025]
Abstract
Low-temperature second near-infrared region (NIR-II) photothermal therapy (PTT) has shown significant potential in minimizing damage to normal tissues and reducing inflammation. However, it still faces challenge of insufficient immune response. Thus, a multifunctional phototheranostic nanoparticle (BDPB/Pt/Fe@P[5]) is developed by co-loading BDPB, CDHPt, and Fe2⁺ with a pH-sensitive lipid DSPE-PEOz2K. The carboxylatopillar[5]arene (CP[5]) used to construct this nanoparticle exhibits strong host-guest recognition with pyridine salts, alleviating aggregation caused quench (ACQ) effect and enhancing the NIR-II emission of the donor-acceptor-donor (D-A-D)-type organic small molecule (BDPB). CP[5] provides suitable vehicles for encapsulating platinum (IV) prodrugs (CDHPt) and Fe2⁺ ions via metal coordination for controllable reactive oxygen species (ROS) release. Under low-intensity NIR-II laser irradiation and an acidic tumor microenvironment, the nanoparticles degrade, releasing CDHPt and Fe2⁺ ions for platinum-based therapy and chemodynamic therapy (CDT). CDHPt facilitates the direct production of superoxide anions (O₂·⁻) from O₂ and partially converts it into the highly cytotoxic hydroxyl radicals, thereby promoting the Fenton reaction process. The therapeutic efficacy is further synergized by immunogenic cell death (ICD) effect.
Collapse
Affiliation(s)
- Miaomiao Ding
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Haoran Chen
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Liuliang He
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhichao Wang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xianghua Zhao
- College of Chemistry and Chemical Engineering, Xinyang Normal University, Xinyang, Henan, 464000, China
| | - Pengfei Sun
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Qunbo Mei
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Daifeng Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Quli Fan
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| |
Collapse
|
14
|
Jung LA, Demir S, Hotes A, Hiyama E, Hishiki T, Indersie E, Branchereau S, Cairo S, Kappler R. Targeting HSP90 with Ganetespib to Induce CDK1 Degradation and Promote Cell Death in Hepatoblastoma. Cancers (Basel) 2025; 17:1341. [PMID: 40282517 PMCID: PMC12026307 DOI: 10.3390/cancers17081341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Hepatoblastoma, the most common malignant liver tumor in pediatric patients, is characterized by a remarkably low mutation rate, thereby impeding targeted therapies. Current treatment regimens rely on conventional cytotoxic agents that often cause severe adverse effects, necessitating the search for novel, less toxic therapeutic approaches. METHODS In this study, we explored the anti-tumor potential of heat shock protein 90 (HSP90) inhibitors using a unique collection of hepatoblastoma in vitro models. RESULTS Among the five tested inhibitors, we identified ganetespib as the most effective, significantly suppressing tumor cell growth while sparing healthy, non-tumor cells. Ganetespib treatment at low nanomolar concentrations markedly reduced cell proliferation, impaired long-term survival, and inhibited three-dimensional spheroid growth, ultimately leading to the induction of apoptosis. Mechanistically, ganetespib downregulated the expression of the HSP90 client protein cyclin-dependent kinase 1, a key cell cycle regulator controlling G2/M phase transition, which is heavily upregulated in hepatoblastoma. This disruption consequently resulted in cell cycle arrest, further contributing to its anti-tumor effects. CONCLUSIONS HSP90 inhibition by ganetespib demonstrates significant potential as a novel therapeutic strategy for hepatoblastoma, offering a potential alternative to current cytotoxic treatments with fewer adverse effects.
Collapse
Affiliation(s)
- Lea Amelie Jung
- Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, LMU University Hospital, 80337 Munich, Germany; (L.A.J.); (S.D.); (A.H.)
| | - Salih Demir
- Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, LMU University Hospital, 80337 Munich, Germany; (L.A.J.); (S.D.); (A.H.)
| | - Alina Hotes
- Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, LMU University Hospital, 80337 Munich, Germany; (L.A.J.); (S.D.); (A.H.)
| | - Eiso Hiyama
- Natural Science Center for Basic Research and Development, Hiroshima University, Hiroshima 739-0046, Japan;
| | - Tomoro Hishiki
- Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8677, Japan;
| | | | | | - Stefano Cairo
- Champions Oncology, Inc., Rockville, MD 20850, USA;
- Pediatric Research Institute (IRP), 35127 Padova, Italy
| | - Roland Kappler
- Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, LMU University Hospital, 80337 Munich, Germany; (L.A.J.); (S.D.); (A.H.)
| |
Collapse
|
15
|
Isermann T, Schneider KL, Wegwitz F, De Oliveira T, Conradi LC, Volk V, Feuerhake F, Papke B, Stintzing S, Mundt B, Kühnel F, Moll UM, Schulz-Heddergott R. Enhancement of colorectal cancer therapy through interruption of the HSF1-HSP90 axis by p53 activation or cell cycle inhibition. Cell Death Differ 2025:10.1038/s41418-025-01502-x. [PMID: 40204953 DOI: 10.1038/s41418-025-01502-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
The stress-associated chaperone system is an actionable target in cancer therapies. It is ubiquitously upregulated in cancer tissues and enables tumorigenicity by stabilizing oncoproteins. Most inhibitors target the key component, heat-shock protein 90 (HSP90). Although HSP90 inhibitors are highly tumor-selective, they fail in clinical trials. These failures are partly due to interference with a negative regulatory feedback loop in the heat-shock response (HSR): in response to HSP90 inhibition, there is compensatory synthesis of stress-inducible chaperones, mediated by the transcription factor heat-shock-factor 1 (HSF1). We recently identified that wild-type p53 reduces the HSR by repressing HSF1 via a p21-CDK4/6-MAPK-HSF1 axis. Here, we test whether in HSP90-based therapies, simultaneous p53 activation or direct cell cycle inhibition interrupts the deleterious HSF1-HSR axis and improves the efficiency of HSP90 inhibitors. We found that the clinically relevant p53 activator Idasanutlin suppresses the HSF1-HSR activity in HSP90 inhibitor-based therapies. This combination synergistically reduces cell viability and accelerates cell death in p53-proficient colorectal cancer (CRC) cells, murine tumor-derived organoids, and patient-derived organoids (PDOs). Mechanistically, upon combination therapy, CRC cells upregulate p53-associated pathways, apoptosis, and inflammatory pathways. Likewise, in a CRC mouse model, dual HSF1-HSP90 inhibition represses tumor growth and remodels immune cell composition. Importantly, inhibition of the cyclin-dependent kinases 4/6 (CDK4/6) under HSP90 inhibition phenocopies synergistic repression of the HSR in p53-proficient CRC cells. Moreover, in p53-deficient CRC cells, HSP90 inhibition in combination with CDK4/6 inhibitors similarly suppresses the HSF1-HSR and reduces cancer growth. Likewise, p53-mutated PDOs respond to dual HSF1-HSP90 inhibition, providing a strategy to target CRC independent of the p53 status. In sum, we provide new options to improve HSP90-based therapies to enhance CRC therapies.
Collapse
Affiliation(s)
- Tamara Isermann
- Department of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
- Laboratory of Molecular Tumor Pathology and Systems Biology, Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK); Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kim Lucia Schneider
- Department of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Florian Wegwitz
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago De Oliveira
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Lena-Christin Conradi
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Valery Volk
- Institute for Pathology, Hannover Medical School, Hannover, Germany
| | | | - Björn Papke
- Laboratory of Molecular Tumor Pathology and Systems Biology, Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK); Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Stintzing
- German Cancer Consortium (DKTK); Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematology, Oncology and Cancer Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Bettina Mundt
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Florian Kühnel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Ute M Moll
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | | |
Collapse
|
16
|
Wu Z, Li Y, Dong J, Qin JJ. An updated review on the role of small molecules in mediating protein degradation. Eur J Med Chem 2025; 287:117370. [PMID: 39933402 DOI: 10.1016/j.ejmech.2025.117370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/25/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Targeted protein degradation (TPD) technologies, inspired by physiological processes, have recently provided new directions for drug development. Unlike conventional drug development focusing on targeting the active sites of disease-related proteins, TPD can utilize any nook or cranny of a protein to drive degradation through the cell's inherent destruction mechanism. It offers various advantages such as stronger pharmacological effects, an expanded range of drug targets, and higher selectivity. Based on the ubiquitin-proteasome system and the lysosomal degradation pathway, a variety of TPD strategies have been developed including PROTAC, PROTAB, and AUTOTAC. These TPD strategies have continuously enriched the toolbox for targeted protein degradation and expanded the scope of application, providing new ideas for biological research and drug discovery. This review attempts to introduce up-to-date research progress in the TPD strategies, focusing mainly on their design concepts, advantages, potential applications, and challenges, which may provide some inspiration for drug design, drug discovery, and clinical application for biologists and chemists.
Collapse
Affiliation(s)
- Zumei Wu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yulong Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jinyun Dong
- Center for Innovative Drug Research, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China.
| | - Jiang-Jiang Qin
- Center for Innovative Drug Research, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China.
| |
Collapse
|
17
|
Muraoka H, Kazuno H, Hashimoto A, Sootome H, Ohkubo S. Pimitespib, an HSP90 Inhibitor, Enhances the Efficacy of PARP Inhibitors in PARP Inhibitor-Insensitive Breast Cancer Cells. Cancer Sci 2025. [PMID: 40167031 DOI: 10.1111/cas.70058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 02/18/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
Heat shock protein 90 (HSP90) plays a crucial role in the maintenance of protein homeostasis in cancer cells. Inhibition of HSP90 is anticipated to exert anticancer activities by reducing levels of HSP90 client proteins. Pimitespib (TAS-116) has emerged as a potent ATP-competitive inhibitor of both HSP90α and β, demonstrating favorable therapeutic properties in preclinical models. Notably, pimitespib is the first HSP90 inhibitor approved for the treatment of advanced gastrointestinal stromal tumors in Japan. Poly(ADP-ribose) polymerase (PARP) inhibitors target cancers susceptible to the homologous recombination (HR) pathway and are used for treating various types of tumors, particularly those harboring defects in HR repair pathways within DNA damage repair (DDR) such as mutations in breast cancer genes 1 and 2 (BRCA1 and BRCA2, respectively). However, PARP inhibitors have shown limited efficacy in HR-proficient tumors, and the development of resistance to PARP inhibitors via restoration of DDR systems poses a significant challenge. In this study, we explored the potential of pimitespib to enhance PARP inhibitor activity. In PARP inhibitor-insensitive breast cancer cell lines, pimitespib impaired HR pathway function by promoting the proteasome-mediated degradation of proteins involved in HR, such as BRCA1, BRCA2, and Rad51 homologous 1 (RAD51). Consequently, pimitespib enhanced antitumor activity and DNA damage induced by PARP inhibitors in vitro. In human breast cancer xenograft mouse models, pimitespib downregulated RAD51 proteins and augmented the antitumor effects of PARP inhibitors. These findings highlight the potential of pimitespib as a therapeutic agent in combination with PARP inhibitors to treat PARP inhibitor-insensitive cancers.
Collapse
Affiliation(s)
- Hiromi Muraoka
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Hiromi Kazuno
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Akihiro Hashimoto
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Hiroshi Sootome
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Shuichi Ohkubo
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| |
Collapse
|
18
|
Rivera D, Bouras A, Mattioli M, Anastasiadou M, Pacentra AC, Pelcher O, Koziel C, Schupper AJ, Chanenchuk T, Carlton H, Ivkov R, Hadjipanayis CG. Magnetic hyperthermia therapy enhances the chemoradiosensitivity of glioblastoma. Sci Rep 2025; 15:10532. [PMID: 40148452 PMCID: PMC11950323 DOI: 10.1038/s41598-025-95544-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/21/2025] [Indexed: 03/29/2025] Open
Abstract
Glioblastoma (GBM) is the most common primary brain cancer and is resistant to standard-of-care chemoradiation therapy (CRT). Magnetic hyperthermia therapy (MHT) exposes magnetic iron oxide nanoparticles (MIONPs) to an alternating magnetic field (AMF) to generate local hyperthermia. This study evaluated MHT-mediated enhancement of CRT in preclinical GBM models. Cell viability and apoptosis were assessed in GBM cell lines after water bath heating with radiation and/or temozolomide. Heating efficiency of MIONPs after intracranial delivery was measured in healthy mice. MHT with CRT was performed in syngeneic and patient-derived xenograft (PDX) GBM tumors. Tissue sections were analyzed for γ-H2AX, HSP90, CD4 + T cells, and microglial cells. Tumor burden and survival were assessed. Hyperthermia with radiation and temozolomide significantly reduced cell viability and increased apoptosis. Hyperthermia predominantly exhibited additive to synergistic interactions with both treatment modalities and reduced doses needed for tumor cell growth inhibition. In vivo, MHT with CRT decreased tumor burden and increased survival in PDX and syngeneic models. Immunohistochemistry showed increased γ-H2AX, HSP90, microglial activation, and CD4 + T cells after MHT in combination with CRT. Overall, adjuvant hyperthermia enhances CRT efficacy in GBM cells, with MHT improving survival outcomes in rodents. Sufficient intracranial heating and MIONP retention for repeated treatments was achieved, supporting further clinical translation.
Collapse
Affiliation(s)
- Daniel Rivera
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Alexandros Bouras
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Milena Mattioli
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Maria Anastasiadou
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Anna Chiara Pacentra
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Olivia Pelcher
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Corrine Koziel
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Alexander J Schupper
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Tori Chanenchuk
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Hayden Carlton
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Constantinos G Hadjipanayis
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA.
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA.
| |
Collapse
|
19
|
Ewert W, Bartens C, Ongouta J, Holmes M, Heutling A, Kishore A, Urbansky T, Zeilinger C, Preller M, Kirschning A. Structure and function of the geldanamycin amide synthase from Streptomyces hygroscopicus. Nat Commun 2025; 16:2464. [PMID: 40075103 PMCID: PMC11903869 DOI: 10.1038/s41467-025-57013-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Amide synthases catalyze the formation of macrolactam rings from aniline-containing polyketide-derived seco-acids as found in the important class of ansamycin antibiotics. One of these amide synthases is the geldanamycin amide synthase GdmF, which we recombinantly expressed, purified and studied in detail both functionally as well as structurally. Here we show that purified GdmF catalyzes the amide formation using synthetically derived substrates. The atomic structures of the ligand-free enzyme and in complex with simplified substrates reveal distinct structural features of the substrate binding site and a putative role of the flexible interdomain region for the catalysis reaction.
Collapse
Affiliation(s)
- Wiebke Ewert
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Christian Bartens
- Institute of Organic Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Jekaterina Ongouta
- Institute of Organic Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Monika Holmes
- Institute of Organic Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Anja Heutling
- Institute of Organic Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Anusha Kishore
- Center of Biomolecular Drug Research (BMWZ) Leibniz University Hannover, Hannover, Germany
| | - Tim Urbansky
- Institute for Functional Gene Analytics (IFGA), University of Applied Sciences Bonn-Rhein-Sieg, Rheinbach, Germany
| | - Carsten Zeilinger
- Center of Biomolecular Drug Research (BMWZ) Leibniz University Hannover, Hannover, Germany.
| | - Matthias Preller
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany.
- Institute for Functional Gene Analytics (IFGA), University of Applied Sciences Bonn-Rhein-Sieg, Rheinbach, Germany.
| | - Andreas Kirschning
- Institute of Organic Chemistry, Leibniz University Hannover, Hannover, Germany.
- Uppsala Biomedical Center (BMC), University Uppsala, Uppsala, Sweden.
| |
Collapse
|
20
|
Gu J, He Y, He C, Zhang Q, Huang Q, Bai S, Wang R, You Q, Wang L. Advances in the structures, mechanisms and targeting of molecular chaperones. Signal Transduct Target Ther 2025; 10:84. [PMID: 40069202 PMCID: PMC11897415 DOI: 10.1038/s41392-025-02166-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/25/2024] [Accepted: 01/15/2025] [Indexed: 03/15/2025] Open
Abstract
Molecular chaperones, a class of complex client regulatory systems, play significant roles in the prevention of protein misfolding and abnormal aggregation, the modulation of protein homeostasis, and the protection of cells from damage under constantly changing environmental conditions. As the understanding of the biological mechanisms of molecular chaperones has increased, their link with the occurrence and progression of disease has suggested that these proteins are promising targets for therapeutic intervention, drawing intensive interest. Here, we review recent advances in determining the structures of molecular chaperones and heat shock protein 90 (HSP90) chaperone system complexes. We also describe the features of molecular chaperones and shed light on the complicated regulatory mechanism that operates through interactions with various co-chaperones in molecular chaperone cycles. In addition, how molecular chaperones affect diseases by regulating pathogenic proteins has been thoroughly analyzed. Furthermore, we focus on molecular chaperones to systematically discuss recent clinical advances and various drug design strategies in the preclinical stage. Recent studies have identified a variety of novel regulatory strategies targeting molecular chaperone systems with compounds that act through different mechanisms from those of traditional inhibitors. Therefore, as more novel design strategies are developed, targeting molecular chaperones will significantly contribute to the discovery of new potential drugs.
Collapse
Affiliation(s)
- Jinying Gu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yanyi He
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chenxi He
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qiuyue Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qifei Huang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shangjun Bai
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ruoning Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.
- Jiangsu Provincial TCM Engineering Technology Research Center of Highly Efficient Drug Delivery Systems (DDSs), Nanjing, China.
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China.
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Lei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China.
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
21
|
Min L, Li X, Liang L, Ruan Z, Yu S. Targeting HSP90 in Gynecologic Cancer: Molecular Mechanisms and Therapeutic Approaches. Cell Biochem Biophys 2025; 83:177-192. [PMID: 39249180 DOI: 10.1007/s12013-024-01502-7] [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] [Accepted: 08/25/2024] [Indexed: 09/10/2024]
Abstract
One of the leading causes of mortality for women is gynecologic cancer (GC). Numerous molecules (tumor suppressor genes or oncogenes) are involved in this form of cancer's invasion, metastasis, tumorigenic process, and therapy resistance. Currently, there is a shortage of efficient methods to eliminate these diseases, hence it is crucial to carry out more extensive studies on GCs. Novel pharmaceuticals are required to surmount this predicament. Highly conserved molecular chaperon, heat shock protein (HSP) 90, is essential for the maturation of recently produced polypeptides and offers a refuge for misfolding or denatured proteins to be turned around. In cancer, the client proteins of HSP90 play a role in the entire process of oncogenesis, which is linked to all the characteristic features of cancer. In this study, we explore the various functions of HSPs in GC progression. We also discuss their potential as promising targets for pharmacological therapy.
Collapse
Affiliation(s)
- Lu Min
- Changchun University of Chinese Medicine Hospital, Changchun, 130000, China
| | - Xuewei Li
- Changchun University of Chinese Medicine Hospital, Changchun, 130000, China
| | - Lily Liang
- Changchun University of Chinese Medicine Hospital, Changchun, 130000, China
| | - Zheng Ruan
- Department of Traditional Chinese Medicine, 964th Hospital, Changchun, 130000, China
| | - Shaohui Yu
- Changchun University of Chinese Medicine Hospital, Changchun, 130000, China.
| |
Collapse
|
22
|
Huang L, Wang W, Zhu Z, Li Q, Li M, Zhou H, Xu Q, Wen W, Wang Q, Yu F. Novel starting points for fragment-based drug design against human heat-shock protein 90 identified using crystallographic fragment screening. IUCRJ 2025; 12:177-187. [PMID: 39819741 PMCID: PMC11878448 DOI: 10.1107/s2052252524012247] [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: 07/26/2024] [Accepted: 12/18/2024] [Indexed: 01/19/2025]
Abstract
Heat-shock protein 90 (HSP90) is a highly active molecular chaperone that plays a crucial role in cellular function. It facilitates the folding, assembly and stability of various oncogenic proteins, particularly kinases and transcription factors involved in regulating tumor growth and maintenance signaling pathways. Consequently, HSP90 inhibitors are being explored as drugs for cancer therapy. Crystallographic fragment screening is a novel screening method that has been developed in recent years for fragment-based drug discovery and is known for its high hit rate and its ability to provide direct insights into the complex structures of proteins and compounds. In this paper, high-diffraction-resolution crystals of the N-terminal domain of human HSP90α were employed in crystallographic fragment screening to discover binding fragments and binding sites. A diverse library of 800 structurally distinct fragments was screened, yielding 91 starting points for the fragment-based drug design of new HSP90α N-terminal inhibitors. Nearly a thousand crystals were measured, with 738 being processed and phased using a highly automated data-processing pipeline including data reduction and phasing, refinement and hit identification via PanDDA multi-data-set analysis. The 91 identified compounds bind to eight distinct regions of the HSP90α N-terminus, with 63 fragments located in the ATP-binding pocket and its surroundings, thus demonstrating the potential for the development of HSP90α- and ATP-binding inhibitors. This study emphasizes crystallographic fragment screening as a powerful method that can effectively identify fragment molecules and inhibitors that bind to HSP90α, contributing to ongoing efforts in cancer drug discovery.
Collapse
Affiliation(s)
- Liqing Huang
- Shanghai Institute of Applied Physics, Chinese Academy of SciencesShanghai201800People’s Republic of China
- University of Chinese Academy of SciencesBeijing100049People’s Republic of China
| | - Weiwei Wang
- Shanghai Synchrotron Radiation FacilityShanghai Advanced Research Institute, Chinese Academy of SciencesShanghai201204People’s Republic of China
| | - Zhimin Zhu
- Shanghai Institute of Applied Physics, Chinese Academy of SciencesShanghai201800People’s Republic of China
- University of Chinese Academy of SciencesBeijing100049People’s Republic of China
| | - Qianhui Li
- Shanghai Institute of Applied Physics, Chinese Academy of SciencesShanghai201800People’s Republic of China
- University of Chinese Academy of SciencesBeijing100049People’s Republic of China
| | - Minjun Li
- Shanghai Synchrotron Radiation FacilityShanghai Advanced Research Institute, Chinese Academy of SciencesShanghai201204People’s Republic of China
| | - Huan Zhou
- Shanghai Institute of Applied Physics, Chinese Academy of SciencesShanghai201800People’s Republic of China
- Shanghai Synchrotron Radiation FacilityShanghai Advanced Research Institute, Chinese Academy of SciencesShanghai201204People’s Republic of China
| | - Qin Xu
- Shanghai Institute of Applied Physics, Chinese Academy of SciencesShanghai201800People’s Republic of China
- Shanghai Synchrotron Radiation FacilityShanghai Advanced Research Institute, Chinese Academy of SciencesShanghai201204People’s Republic of China
| | - Wen Wen
- Shanghai Institute of Applied Physics, Chinese Academy of SciencesShanghai201800People’s Republic of China
- Shanghai Synchrotron Radiation FacilityShanghai Advanced Research Institute, Chinese Academy of SciencesShanghai201204People’s Republic of China
| | - Qisheng Wang
- Shanghai Institute of Applied Physics, Chinese Academy of SciencesShanghai201800People’s Republic of China
- Shanghai Synchrotron Radiation FacilityShanghai Advanced Research Institute, Chinese Academy of SciencesShanghai201204People’s Republic of China
| | - Feng Yu
- Shanghai Institute of Applied Physics, Chinese Academy of SciencesShanghai201800People’s Republic of China
- Shanghai Synchrotron Radiation FacilityShanghai Advanced Research Institute, Chinese Academy of SciencesShanghai201204People’s Republic of China
| |
Collapse
|
23
|
Tran C, Hamze A. Recent Advancements in the Development of HDAC/Tubulin Dual-Targeting Inhibitors. Pharmaceuticals (Basel) 2025; 18:341. [PMID: 40143119 PMCID: PMC11945613 DOI: 10.3390/ph18030341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
Histone deacetylases (HDACs) have become one of the main targets in cancer therapy due to their involvement in various biological processes, including gene regulation, cell proliferation, and differentiation. Microtubules, as key elements of the cell cytoskeleton, also represent important therapeutic targets in anticancer drugs research. These proteins are involved in diverse cellular functions, especially mitosis, cell signaling, and intracellular trafficking. With the emergence of multi-target therapy during the last decades, the combination of HDAC and tubulin inhibitors has been envisioned as a practical approach for optimizing the therapeutic efficacy of antitumor molecules. HDAC/tubulin dual-targeting inhibitors offer the advantages of the synergistic action of both compounds, along with a significant decrease in their respective toxicities and drug resistance. This review will detail the major recent advancements in the development of HDAC/tubulin dual inhibitors over the last decade and their impact on anticancer drugs discovery.
Collapse
Affiliation(s)
- Christine Tran
- BioCIS, CNRS (Centre National de Recherche Scientifique), Université Paris-Saclay, 91400 Orsay, France
| | - Abdallah Hamze
- BioCIS, CNRS (Centre National de Recherche Scientifique), Université Paris-Saclay, 91400 Orsay, France
| |
Collapse
|
24
|
Narykina V, Kleynhans J, Cawthorne C, Schymkowitz J, Rousseau F, Bormans G. Development and evaluation of Hsp90-targeting nanobodies for visualisation of extracellular Hsp90 in tumours using PET imaging. EJNMMI Radiopharm Chem 2025; 10:10. [PMID: 39982615 PMCID: PMC11845643 DOI: 10.1186/s41181-025-00331-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/05/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND The extracellular localisation of the Heat shock protein 90 (Hsp90) is associated with the diseased state and wound healing and presents a promising opportunity for cancer targeting using Positron Emission Tomography (PET) imaging and molecularly targeted radiotherapy. The aim of this work is to develop a radiotracer with low nanomolar binding affinity to target the extracellular and particularly membrane pool of Hsp90, evaluate it in vitro, and conduct preliminary PET studies in vivo in mouse tumour models. Variable Heavy domain of Heavy chain antibodies, often referred to as Nanobodies, are suitable targeting vectors for the extracellular targets due to their favourable pharmacokinetic properties and low nanomolar target affinities. The main objective of the study is to target tumours expressing extracellular and membrane Hsp90 phenotype with minimal tracer accumulation in the non-target organs, which limited the translation of previously studied small molecule cytosolic Hsp90 tracers suffering from high non-Hsp90 specific background in the abdominal area. RESULTS Six nanobodies were obtained after llama immunization with recombinant Hsp90α and ELISA biopanning, produced in E. coli and screened for stability and affinity. We selected one nanobody, 4DAM26, with good thermal stability, no aggregation at elevated temperatures, and low nanomolar affinity towards Hsp90α and Hsp90β isoforms for translation as a PET radiotracer. The nanobody was bioconjugated to p-NCS-NODAGA and radiolabeled with gallium-68 with 75 ± 11% radiochemical yield and > 99% radiochemical purity and remained stable up to 3 h in phosphate buffered saline and mouse serum. Pilot in vivo evaluation using µPET/CT and ex vivo biodistribution demonstrated a favourable pharmacokinetic profile, but the tumour uptake was non-distinguishable from the background tissue. CONCLUSION Compared to the small molecule Hsp90 tracers, the studied Nb-based tracer has improved pharmacokinetics properties including renal clearance and almost no accumulation in the non-target organs. Tumour uptake, on the other hand, was minimal and could not be differentiated from the background in µPET/CT. Our experiments indicate that in the studied models, membrane and extracellular expression of Hsp90 is majorly an artifact of cellular death, as only dead/dying cells had accessible pools of Hsp90 by flow cytometry, a consequence of a leaky membrane. More fundamental research is required to reassess the role of extracellular Hsp90 in cancer, and our future efforts will be focused on improving our inventory of cytosolic Hsp90 tracers with proven Hsp90-specific tumour accumulation.
Collapse
Affiliation(s)
- Valeria Narykina
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000, Leuven, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
- Switch Laboratory, VIB Center for Brain and Disease Research, Herestraat 49, 3000, Leuven, Belgium
| | - Janke Kleynhans
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Christopher Cawthorne
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, University of Leuven, 3000, Leuven, Belgium
| | - Joost Schymkowitz
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
- Switch Laboratory, VIB Center for Brain and Disease Research, Herestraat 49, 3000, Leuven, Belgium
| | - Frederic Rousseau
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
- Switch Laboratory, VIB Center for Brain and Disease Research, Herestraat 49, 3000, Leuven, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000, Leuven, Belgium.
| |
Collapse
|
25
|
Yao M, Yan W, Wang Y, Zhao Y, Xu X, Chen Y, Yu C, Li Y, Jiang H, Shen J, Cheng J, Xie C. IHCH9033, a novel class I HDAC inhibitor, synergizes with FLT3 inhibitor and rescues quizartinib resistance in FLT3-ITD AML via enhancing DNA damage response. Exp Hematol Oncol 2025; 14:15. [PMID: 39955584 PMCID: PMC11829435 DOI: 10.1186/s40164-025-00605-y] [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: 11/26/2024] [Accepted: 01/30/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Despite initial success with FLT3 inhibitors (FLT3is), outcomes for FLT3-ITD acute myeloid leukemia (AML) patients remain unsatisfactory, underscoring the need for more effective treatment options. Epigenetic modifications, such as histone acetylation, contribute to AML's onset and persistence, advocating the potential for epigenetic therapies. However, the poor specificity of pan-histone deacetylase inhibitors (HDACis) leads to undesirable adverse effects, prompting the need for isoform-specific HDACis. This study aims to explore the antileukemic activities and mechanisms of IHCH9033, a novel class I HDACi, alone or combined with FLT3i in FLT3-ITD AML. METHODS The viability of AML cell lines and primary AML cells treated with HDACis alone or in combination with FLT3i was detected by MTT or CCK8 assay. Flow cytometry was utilized to examine cell apoptosis, cell cycle progression and ROS production. RNA sequencing analysis, RT-qPCR, western blotting, and co-immunoprecipitation assays were employed to elucidate the molecule mechanisms. The in vivo anti-leukemia efficacy was tested in xenografted mice models derived from FLT3-ITD cell lines and primary AML patients. RESULTS Here, we identified IHCH9033, a novel selective class I HDACi, which exhibited an increased antitumor effect in FLT3-ITD AML through effectively eliminating leukemia burden and overcoming resistance to FLT3i. Mechanically, IHCH9033 selectively inhibited DNA repair in FLT3-ITD AML cells, leading to the accumulation of DNA damage that eventually resulted in cell cycle arrest and apoptosis. Additionally, IHCH9033 induced HSP90 acetylation, FLT3 ubiquitination, and proteasomal degradation of FLT3, thereby inhibiting FLT3 downstream signaling. Notably, IHCH9033 maintained its potency in both FLT3i-resistant AML cell lines and primary-resistant patient samples, and exerted strong synergy with the FLT3i quizartinib, leading to tumor regression in FLT3-ITD/TKD AML xenografts. In patient-derived xenografts, the treatment with IHCH9033, both alone and in combination, led to nearly complete eradication of the AML burden, without significant adverse effects. CONCLUSIONS Our study shows that IHCH9033, a novel class I HDACi with a desirable pharmacological profile, is a promising drug candidate for FLT3-ITD AML, and suggests a strategy of combining class I HDACis and FLT3is in AML clinical trials to increase efficacy and overcome resistance, thus potentially providing a curative treatment option.
Collapse
Affiliation(s)
- Mingyue Yao
- Lingang Laboratory, 2380 Hechuan Road, Shanghai, 201101, China
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Wenzhong Yan
- iHuman Institute, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai, 201210, China
| | - Yafang Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Yu Zhao
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Xiaowei Xu
- Department of Hematology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai General Hospital, Shanghai, 200025, China
| | - Yujun Chen
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Chengcheng Yu
- Lingang Laboratory, 2380 Hechuan Road, Shanghai, 201101, China
| | - Yingnian Li
- Lingang Laboratory, 2380 Hechuan Road, Shanghai, 201101, China
| | - Hualiang Jiang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Drug Discovery and Development Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jie Shen
- Department of Pharmacy, The SATCM Third Grade Laboratory of Traditional Chinese Medicine Preparations, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Shanghai, 201203, China.
| | - Jianjun Cheng
- iHuman Institute, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai, 201210, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Chengying Xie
- Lingang Laboratory, 2380 Hechuan Road, Shanghai, 201101, China.
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
26
|
Smadja DM, Abreu MM. Hyperthermia and targeting heat shock proteins: innovative approaches for neurodegenerative disorders and Long COVID. Front Neurosci 2025; 19:1475376. [PMID: 39967803 PMCID: PMC11832498 DOI: 10.3389/fnins.2025.1475376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 01/03/2025] [Indexed: 02/20/2025] Open
Abstract
Neurodegenerative diseases (NDs) and Long COVID represent critical and growing global health challenges, characterized by complex pathophysiological mechanisms including neuronal deterioration, protein misfolding, and persistent neuroinflammation. The emergence of innovative therapeutic approaches, such as whole-body hyperthermia (WBH), offers promising potential to modulate underlying pathophysiological mechanisms in NDs and related conditions like Long COVID. WBH, particularly in fever-range, enhances mitochondrial function, induces heat shock proteins (HSPs), and modulates neuroinflammation-benefits that pharmacological treatments often struggle to replicate. HSPs such as HSP70 and HSP90 play pivotal roles in protein folding, aggregation prevention, and cellular protection, directly targeting pathological processes seen in NDs like Alzheimer's, Parkinson's, and Huntington's disease. Preliminary findings also suggest WBH's potential to alleviate neurological symptoms in Long COVID, where persistent neuroinflammation and serotonin dysregulation are prominent. Despite the absence of robust clinical trials, the therapeutic implications of WBH extend to immune modulation and the restoration of disrupted physiological pathways. However, the dual nature of hyperthermia's effects-balancing pro-inflammatory and anti-inflammatory responses-emphasizes the need for dose-controlled applications and stringent patient monitoring to minimize risks in vulnerable populations. While WBH shows potential interest, significant challenges remain. These include individual variability in response, limited accessibility to advanced hyperthermia technologies, and the need for standardized clinical protocols. Future research must focus on targeted clinical trials, biomarker identification, and personalized treatment strategies to optimize WBH's efficacy in NDs and Long COVID. The integration of WBH into therapeutic paradigms could mark a transformative step in addressing these complex conditions.
Collapse
Affiliation(s)
- David M. Smadja
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, Team Endotheliopathy and Hemostasis Disorders, Paris, France
- Hematology Department, Hôpital Européen Georges Pompidou, Assistance Publique Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Paris, France
| | - M. Marc Abreu
- BTT Medical Institute, Aventura, FL, United States
- BTT Engineering Department, Aventura, FL, United States
| |
Collapse
|
27
|
Feng Z, Liu H, Huang J, Tan X, Su C, Mo S, Tang J, Wang H, Lin Y, Zhang L. Expression of extracellular HSP90α in pre- and post-treatment samples of driver-gene-negative non-small cell lung cancer. Int Immunopharmacol 2025; 146:113792. [PMID: 39689601 DOI: 10.1016/j.intimp.2024.113792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 10/22/2024] [Accepted: 12/03/2024] [Indexed: 12/19/2024]
Abstract
PURPOSE This study aimed to explore the significance of the expression of pre- and post-treatment eHSP90α in the treatment response evaluation and prognosis of driver-gene-negative non-small cell lung cancer (NSCLC), as well as the significance of eHSP90α expression in the prognosis of immunotherapy. METHODS We collected pre-treatment eHSP90α in 330 driver-gene-negative NSCLC patients and analyzed its relationship with efficacy evaluation and prognosis. Survival curves were used to determine their respective critical values and the relationship between eHSP90α expression and OS and PFS was analyzed. Then, univariable and multivariable Cox regression analyses and LASSO multivariable logistic regression analyses were used to establish the prognostic and efficacy evaluation models. RESULTS High expression of pre-treatment eHSP90α in driver-gene-negative NSCLC patients is associated with shorter OS. Pre-treatment eHSP90α, immunotherapy, TTF-1, CA125, and age influence OS in NSCLC patients. In the stratified analysis of immunotherapy, pre-treatment eHSP90α in immunotherapy is an independent influencing factor of OS in NSCLC patients, and pre-treatment eHSP90α, age, CA125, and B lymphocytes in non-immunotherapy are independent influencing factors of OS in NSCLC patients. High expression of pre-treatment eHSP90α in NSCLC patients is associated with shorter PFS. Pre-treatment eHSP90α, M1, Ki-67, and immunotherapy are independent influencing factors of PFS in NSCLC patients. In the stratified analysis of immunotherapy, pre-treatment eHSP90α and CEA in immunotherapy are independent influencing factors of PFS in NSCLC patients, and pre-treatment eHSP90α, M1, Ki-67, CA125, Th/Ts, and age in non-immunotherapy are independent factors affecting PFS in NSCLC patients. Pre-treatment eHSP90α was one of the influencing factors in the efficacy evaluation model. CONCLUSIONS The expression of pre-treatment eHSP90α has significant significance for the prognosis and efficacy evaluation of driver-gene-negative NSCLC.
Collapse
Affiliation(s)
- Zhaorong Feng
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Haizhou Liu
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China; Guangxi Cancer Molecular Medicine Engineering Research Center, Nanning, China; Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Nanning, China
| | - Jinglei Huang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiaoyu Tan
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Chenglin Su
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Shanli Mo
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jiaying Tang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Haiguang Wang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yanyan Lin
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Litu Zhang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China; Guangxi Cancer Molecular Medicine Engineering Research Center, Nanning, China; Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Nanning, China.
| |
Collapse
|
28
|
Meng X, Wu Q, Cao C, Yang W, Chu S, Guo H, Qi S, Bai J. A novel peptide encoded by circSRCAP confers resistance to enzalutamide by inhibiting the ubiquitin-dependent degradation of AR-V7 in castration-resistant prostate cancer. J Transl Med 2025; 23:108. [PMID: 39844192 PMCID: PMC11755828 DOI: 10.1186/s12967-025-06115-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND The sustained activation of androgen receptor splice variant-7 (AR-V7) is a key factor in the resistance of castration-resistant prostate cancer (CRPC) to second-generation anti-androgens such as enzalutamide (ENZ). The AR/AR-V7 protein is regulated by the E3 ubiquitin ligase STUB1 and a complex involving HSP70, but the precise mechanism remains unclear. METHODS High-throughput RNA sequencing was used to identify differentially expressed circular RNAs (circRNAs) in ENZ-resistant and control CRPC cells. The coding potential of circSRCAP was confirmed by polysome profiling and LC-MS. The function of circSRCAP was validated in vitro and in vivo using gain- and loss-of-function assays. Mechanistic insights were obtained through immunoprecipitation analyses. RESULTS A novel ENZ-resistant circRNA, circSRCAP, was identified and shown to be upregulated in ENZ-resistant C4-2B (ENZR-C4-2B) cells, correlating with increased AR-V7 protein levels. circSRCAP is generated via splicing by eIF4A3, forming a loop structure and is exported from the nucleus by the RNA helicase DDX39A. Mechanistically, circSRCAP encodes a 75-amino acid peptide (circSRCAP-75aa) that inhibits the ubiquitination of AR/AR-V7's co-chaperone protein HSP70 by disrupting the interaction with the E3 ligase STUB1. This process results in the upregulation of AR-V7 expression and promotes ENZ resistance in CRPC cells. Xenograft tumor models further confirmed the role of circSRCAP in CRPC progression and its potential as a therapeutic target for ENZ-resistant CRPC. CONCLUSIONS circSRCAP provides an epigenetic mechanism influencing AR-V7 stability and offers a promising therapeutic target for treating ENZ-resistant CRPC.
Collapse
MESH Headings
- Male
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/pathology
- Humans
- Phenylthiohydantoin/pharmacology
- Phenylthiohydantoin/analogs & derivatives
- Phenylthiohydantoin/therapeutic use
- RNA, Circular/metabolism
- RNA, Circular/genetics
- Benzamides
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/drug effects
- Receptors, Androgen/metabolism
- Receptors, Androgen/genetics
- Proteolysis/drug effects
- Cell Line, Tumor
- Animals
- Nitriles
- Ubiquitin/metabolism
- Peptides/chemistry
- Peptides/pharmacology
- Peptides/metabolism
- Peptides/genetics
- Ubiquitin-Protein Ligases/metabolism
- Mice, Nude
- Protein Isoforms/metabolism
- Mice
Collapse
Affiliation(s)
- Xiannan Meng
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Qingxuan Wu
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Chengsong Cao
- Department of Oncology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
- Department of Oncology, Xuzhou Institute of Medical Science, Xuzhou, Jiangsu, China
| | - Wendong Yang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Sufang Chu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Hongjun Guo
- Department of General Surgery, Xi'an Central Hospital, Xi'an, 710004, Shaanxi, China.
| | - Suhua Qi
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China.
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China.
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
29
|
Chen Y, Hu J, Zhang Y, Peng L, Li X, Li C, Wu X, Wang C. Epilepsy therapy beyond neurons: unveiling astrocytes as cellular targets. Neural Regen Res 2025; 21:01300535-990000000-00659. [PMID: 39819836 PMCID: PMC12094549 DOI: 10.4103/nrr.nrr-d-24-01035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 10/16/2024] [Accepted: 12/10/2024] [Indexed: 01/19/2025] Open
Abstract
ABSTRACT Epilepsy is a leading cause of disability and mortality worldwide. However, despite the availability of more than 20 antiseizure medications, more than one-third of patients continue to experience seizures. Given the urgent need to explore new treatment strategies for epilepsy, recent research has highlighted the potential of targeting gliosis, metabolic disturbances, and neural circuit abnormalities as therapeutic strategies. Astrocytes, the largest group of nonneuronal cells in the central nervous system, play several crucial roles in maintaining ionic and energy metabolic homeostasis in neurons, regulating neurotransmitter levels, and modulating synaptic plasticity. This article briefly reviews the critical role of astrocytes in maintaining balance within the central nervous system. Building on previous research, we discuss how astrocyte dysfunction contributes to the onset and progression of epilepsy through four key aspects: the imbalance between excitatory and inhibitory neuronal signaling, dysregulation of metabolic homeostasis in the neuronal microenvironment, neuroinflammation, and the formation of abnormal neural circuits. We summarize relevant basic research conducted over the past 5 years that has focused on modulating astrocytes as a therapeutic approach for epilepsy. We categorize the therapeutic targets proposed by these studies into four areas: restoration of the excitation-inhibition balance, reestablishment of metabolic homeostasis, modulation of immune and inflammatory responses, and reconstruction of abnormal neural circuits. These targets correspond to the pathophysiological mechanisms by which astrocytes contribute to epilepsy. Additionally, we need to consider the potential challenges and limitations of translating these identified therapeutic targets into clinical treatments. These limitations arise from interspecies differences between humans and animal models, as well as the complex comorbidities associated with epilepsy in humans. We also highlight valuable future research directions worth exploring in the treatment of epilepsy and the regulation of astrocytes, such as gene therapy and imaging strategies. The findings presented in this review may help open new therapeutic avenues for patients with drug-resistant epilepsy and for those suffering from other central nervous system disorders associated with astrocytic dysfunction.
Collapse
Affiliation(s)
- Yuncan Chen
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiayi Hu
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Zhang
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lulu Peng
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoyu Li
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cong Li
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xunyi Wu
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Cong Wang
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Shanghai, China
- Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| |
Collapse
|
30
|
Zhang L, Liu W, Zheng Z, Zhang Q, He Y, Gu J, Wang D, Shu H, Yu J, Liu J, Yin X, Zhang L, Zhang J, You Q, Wang L. Allosteric CDC37 Inhibitor Disrupts Chaperone Complex to Block CDK4/6 Maturation. Angew Chem Int Ed Engl 2025; 64:e202413618. [PMID: 39582167 DOI: 10.1002/anie.202413618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/09/2024] [Accepted: 11/22/2024] [Indexed: 11/26/2024]
Abstract
Cell division cycle 37 (CDC37) is a member of the molecular chaperone family and acts as a cochaperone of heat shock protein 90 (HSP90), which is overexpressed in many cancer types as a regulator of protein kinase maturation. In this process, CDC37 selectively recognizes and stabilizes protein kinases by forming a HSP90-CDC37-kinase chaperone complex. The protein-protein interactions (PPIs) of HSP90-CDC37 and CDC37-kinase complexes contribute to malignant tumors, as oncogenic kinases in malignant cells depend upon CDC37 expression. Thus, inhibiting CDC37 to disrupt HSP90-CDC37-kinase chaperone complex reveals as a promising way to achieve selective inhibition of oncogenic kinase maturation. Herein, we report a small-molecule CDC37 inhibitor called DDO-6079 that simultaneously inhibits HSP90-CDC37 and CDC37-CDK4/6 chaperone complex by binding to an allosteric site on CDC37. DDO-6079 selectively inhibited the maturation of multiple oncogenic kinases to escape heat shock response (HSR). Furthermore, DDO-6079 decreased the thermostability of CDK6, reversed the resistance of CDK6 to palbociclib (a successful CDK4/6 inhibitor) in colorectal cancer cells and exhibited efficacy in vivo. Together, the results revealed that DDO-6079 is a first-in-class small molecule CDC37 inhibitor that disrupts the HSP90-CDC37-kinase chaperone complex and provides a new way to block kinase maturation.
Collapse
Affiliation(s)
- Lixiao Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Wei Liu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhen Zheng
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200025, China
- Medicinal Bioinformatics Center, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200025, China
| | - Qiuyue Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yanyi He
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Jinying Gu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Danni Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Huangliang Shu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Jia Yu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Jianfeng Liu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xingyu Yin
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Lianshan Zhang
- Shanghai Hengrui Pharmaceutical Co., Ltd., Shanghai, 200245, China
| | - Jian Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200025, China
- Medicinal Bioinformatics Center, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200025, China
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Lei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
31
|
Zhang J, Yao M, Xia S, Zeng F, Liu Q. Systematic and comprehensive insights into HIF-1 stabilization under normoxic conditions: implications for cellular adaptation and therapeutic strategies in cancer. Cell Mol Biol Lett 2025; 30:2. [PMID: 39757165 DOI: 10.1186/s11658-024-00682-7] [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: 09/27/2024] [Accepted: 12/19/2024] [Indexed: 01/07/2025] Open
Abstract
Hypoxia-inducible factors (HIFs) are essential transcription factors that orchestrate cellular responses to oxygen deprivation. HIF-1α, as an unstable subunit of HIF-1, is usually hydroxylated by prolyl hydroxylase domain enzymes under normoxic conditions, leading to ubiquitination and proteasomal degradation, thereby keeping low levels. Instead of hypoxia, sometimes even in normoxia, HIF-1α translocates into the nucleus, dimerizes with HIF-1β to generate HIF-1, and then activates genes involved in adaptive responses such as angiogenesis, metabolic reprogramming, and cellular survival, which presents new challenges and insights into its role in cellular processes. Thus, the review delves into the mechanisms by which HIF-1 maintains its stability under normoxia including but not limited to giving insights into transcriptional, translational, as well as posttranslational regulation to underscore the pivotal role of HIF-1 in cellular adaptation and malignancy. Moreover, HIF-1 is extensively involved in cancer and cardiovascular diseases and potentially serves as a bridge between them. An overview of HIF-1-related drugs that are approved or in clinical trials is summarized, highlighting their potential capacity for targeting HIF-1 in cancer and cardiovascular toxicity related to cancer treatment. The review provides a comprehensive insight into HIF-1's regulatory mechanism and paves the way for future research and therapeutic development.
Collapse
Affiliation(s)
- Jiayi Zhang
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, 646000, China
| | - Mingxuan Yao
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Shiting Xia
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
| | - Fancai Zeng
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China.
| | - Qiuyu Liu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
32
|
Monteleone G, Cameli P, Bonella F. The role of heat shock protein 90 in idiopathic pulmonary fibrosis: state of the art. Eur Respir Rev 2025; 34:240147. [PMID: 40107664 PMCID: PMC11920893 DOI: 10.1183/16000617.0147-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 12/16/2024] [Indexed: 03/22/2025] Open
Abstract
Heat shock protein 90 (HSP 90) and its isoforms are a group of homodimeric proteins that regulate several cellular processes, such as the elimination of misfolded proteins, cell development and post-translational modifications of kinase proteins and receptors. Due to its involvement in extracellular matrix (ECM) remodelling, myofibroblast differentiation and apoptosis, HSP 90 has been investigated as a key player in the pathogenesis of lung fibrosis. Idiopathic pulmonary fibrosis (IPF) is the most common and deadly interstitial lung disease, due to the progressive distortion of lung parenchyma related to the overproduction and deposition of altered ECM, driven by transforming growth factor-β (TGF-β) dependent and independent pathways. The inhibition or induction of HSP 90 is associated with a reduced or increased expression of TGF-β receptors, respectively, suggesting a role for HSP 90 as a biomarker and therapeutic target in IPF. Experimental drugs such as geldanamycin and its derivatives 17-AAG (17-N-allylamino-17-demethoxygeldanamicin) and 17-DMAG (17-dimethylaminoethylamino-17-demethoxigeldanamycin), along with AUY-922, 1G6-D7, AT-13387, TAS-116 and myricetin, have been found to reduce lung fibrosis in both in vivo and in vitro models, supporting the role of this emerging target. This review aims to illustrate the structure and biological function of HSP 90 in the context of IPF pathobiology, as well as perspective application of this molecule as a biomarker and therapeutic target for IPF.
Collapse
Affiliation(s)
- Giorgio Monteleone
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of Sacred Heart, Rome, Italy
| | - Paolo Cameli
- Respiratory Diseases Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Francesco Bonella
- Center for interstitial and rare lung diseases, Pneumology Department, Ruhrlandklinik University Hospital, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
33
|
Deng Z, Liu L, Xie G, Zheng Z, Li J, Tan W, Deng Y, Zhang J, Liang M, Wu Y, Zhou Z, Li Y, Chen Y, Huang Y, Su H, Wu G, Shi X, Cen S, Liao Y, Liu Y, Zou F, Chen X. Hsp90α promotes lipogenesis by stabilizing FASN and promoting FASN transcription via LXRα in hepatocellular carcinoma. J Lipid Res 2025; 66:100721. [PMID: 39645039 PMCID: PMC11745951 DOI: 10.1016/j.jlr.2024.100721] [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: 03/22/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024] Open
Abstract
Excessive lipid accumulation promotes the occurrence and progression of hepatocellular carcinoma (HCC), accompanied by high levels of fatty acid synthetase (FASN) and more active lipogenesis. Heat shock protein 90 (Hsp90) acts as a chaperone to maintain the stability and activity of the client proteins. Studies have revealed that Hsp90 regulates the lipid metabolism of HCC, but the effect of Hsp90 on FASN still remains unknown. This study aims to discover the mechanism of Hsp90 inhibition on lipid accumulation and investigate the different effects of Hsp90 N-terminal domain inhibitor STA9090 and C-terminal domain inhibitor novobiocin on FASN protein stability and transcription pathway in HCC. We found that HCC cells tended to store lipids, which could be disrupted by Hsp90 inhibitors in vivo and in vitro. High levels of Hsp90α and FASN in tumor tissue had correlation with poor prognosis of HCC patients, and Hsp90α interacted with FASN to maintain its protein stability. Furthermore, N-terminal domain of Hsp90α was essential for process of sterol regulatory element binding protein 1 to activate FASN transcription and Hsp90α prevented proteasomal degradation of liver X receptor α to upregulate FASN transcription via liver X receptor α/sterol regulatory element binding protein 1 axis. Our data reveal that Hsp90α promotes lipid accumulation by increasing the protein stability and FASN mRNA transcription, and can be alleviated by Hsp90 inhibitors, which provides a theoretical basis for Hsp90-targeted therapy on lipid metabolism in HCC.
Collapse
Affiliation(s)
- Zihao Deng
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Lixia Liu
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Guantai Xie
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhenming Zheng
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jieyou Li
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wenchong Tan
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yaotang Deng
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jinxin Zhang
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Manfeng Liang
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yingxia Wu
- Department of Hygiene Inspection and Quarantine Science, School of Public Health, Southern Medical University, Guangdong Provincial Key Laboratory of Tropical Disease Research, Guangzhou, China
| | - Zhifeng Zhou
- Department of Hygiene Inspection and Quarantine Science, School of Public Health, Southern Medical University, Guangdong Provincial Key Laboratory of Tropical Disease Research, Guangzhou, China
| | - Yan Li
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yukui Chen
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yaling Huang
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Hairou Su
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Guibing Wu
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiongjie Shi
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Shengpei Cen
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yandan Liao
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yilin Liu
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Fei Zou
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xuemei Chen
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China.
| |
Collapse
|
34
|
Fan X, Sun L, Qin Y, Liu Y, Wu S, Du L. The Role of HSP90 Molecular Chaperones in Depression: Potential Mechanisms. Mol Neurobiol 2025; 62:708-717. [PMID: 38896156 DOI: 10.1007/s12035-024-04284-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 06/05/2024] [Indexed: 06/21/2024]
Abstract
Major depressive disorder (MDD) is characterized by high rates of disability and death and has become a public health problem that threatens human life and health worldwide. HPA axis disorder and neuroinflammation are two common biological abnormalities in MDD patients. Hsp90 is an important molecular chaperone that is widely distributed in the organism. Hsp90 binds to the co-chaperone and goes through a molecular chaperone cycle to complete its regulation of the client protein. Numerous studies have demonstrated that Hsp90 regulates how the HPA axis reacts to stress and how GR, the HPA axis' responsive substrate, matures. In addition, Hsp90 exhibits pro-inflammatory effects that are closely related to neuroinflammation in MDD. Currently, Hsp90 inhibitors have made some progress in the treatment of a variety of human diseases, but they still need to be improved. Further insight into the role of Hsp90 in MDD provides new ideas for the development of new antidepressant drugs targeting Hsp90.
Collapse
Affiliation(s)
- Xuyuan Fan
- Department of Medicine, Yangzhou University, Yangzhou, 225012, Jiangsu, China
| | - Lei Sun
- Department of Medicine, Yangzhou University, Yangzhou, 225012, Jiangsu, China
| | - Ye Qin
- Department of Laboratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, Jiangsu, China
| | - Yuan Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, Jiangsu, China
| | - Shusheng Wu
- Department of the Central Laboratory, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, Jiangsu, China.
| | - Longfei Du
- Department of Laboratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, Jiangsu, China.
| |
Collapse
|
35
|
Amatya E, Subramanian C, Long R, McNamara K, Cohen MS, Blagg BSJ. The Investigation of Hsp90C-Terminal Inhibitors Containing Amide Bioisosteres. ChemMedChem 2024; 19:e202400418. [PMID: 39153203 PMCID: PMC11649481 DOI: 10.1002/cmdc.202400418] [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: 05/30/2024] [Revised: 08/15/2024] [Accepted: 08/15/2024] [Indexed: 08/19/2024]
Abstract
Heat Shock Protein 90 (Hsp90) is responsible for the proper folding and maturation of ~400 client protein substrates, many of which are directly associated with the ten hallmarks of cancer. Hsp90 is a great target for cancer therapy including melanoma, since Hsp90 inhibition can disrupt multiple oncogenic pathways simultaneously. In this study, we report the synthesis and anti-proliferative activity manifested by a series of Hsp90 C-terminal inhibitors against mutant BRAF and wild-type BRAF melanoma cells. Furthermore, we explored structure-activity relationships (SAR) for the amide moiety of 6 (B1), a novel Hsp90C-terminal inhibitor via introduction of amide bioisosteres. Compound 6 displayed an IC50 of 1.01 μM, 0.782 μM, 0.607 μM and 1.413 μM against SKMel173, SKMel103, SKMel19 and A375 cells, respectively.
Collapse
Affiliation(s)
- Eva Amatya
- Department of Chemistry and Biochemistry, Warren Family Research Center for Drug Discovery and Development, University of Notre Dame, Notre Dame, Indiana, 46556, USA
| | - Chitra Subramanian
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, Illinois, 61801, United States
| | - Reagan Long
- Department of Chemistry and Biochemistry, Warren Family Research Center for Drug Discovery and Development, University of Notre Dame, Notre Dame, Indiana, 46556, USA
| | - Kelli McNamara
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, Illinois, 61801, United States
| | - Mark S Cohen
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, Illinois, 61801, United States
| | - Brian S J Blagg
- Department of Chemistry and Biochemistry, Warren Family Research Center for Drug Discovery and Development, University of Notre Dame, Notre Dame, Indiana, 46556, USA
| |
Collapse
|
36
|
Dernovšek J, Goričan T, Gedgaudas M, Zajec Ž, Urbančič D, Jug A, Skok Ž, Sturtzel C, Distel M, Grdadolnik SG, Babu K, Panchamatia A, Stachowski TR, Fischer M, Ilaš J, Zubrienė A, Matulis D, Zidar N, Tomašič T. Hiding in plain sight: Optimizing topoisomerase IIα inhibitors into Hsp90β selective binders. Eur J Med Chem 2024; 280:116934. [PMID: 39388906 DOI: 10.1016/j.ejmech.2024.116934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/02/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
Due to their impact on several oncogenic client proteins, the Hsp90 family of chaperones has been widely studied for the development of potential anticancer agents. Although several Hsp90 inhibitors have entered clinical trials, most were unsuccessful because they induced a heat shock response (HSR). This issue can be circumvented by using isoform-selective inhibitors, but the high similarity in the ATP-binding sites between the isoforms presents a challenge. Given that Hsp90 shares a conserved Bergerat fold with bacterial DNA gyrase B and human topoisomerase IIα, we repurposed our ATP-competitive inhibitors of these two proteins for Hsp90 inhibition. We virtually screened a library of in-house inhibitors and identified eleven hits for evaluation of Hsp90 binding. Among these, compound 11 displayed low micromolar affinity for Hsp90 and demonstrated a 12-fold selectivity for Hsp90β over its closest isoform, Hsp90α. Out of 29 prepared analogs, 16 showed a preference for Hsp90β over Hsp90α. Furthermore, eleven of these compounds inhibited the growth of several cancer cell lines in vitro. Notably, compound 24e reduced intracellular levels of Hsp90 client proteins in MCF-7 cells, leading to cell cycle arrest in the G0/G1 phase without inducing HSR. This inhibitor exhibited at least a 27-fold preference for Hsp90β and was selective against topoisomerase IIα, a panel of 22 representative protein kinases, and proved to be non-toxic in a zebrafish larvae toxicology model. Finally, molecular modeling, corroborated by STD NMR studies, and the binding of 24e to the S52A mutant of Hsp90α confirmed that the serine to alanine switch drives the selectivity between the two cytoplasmic isoforms.
Collapse
Affiliation(s)
- Jaka Dernovšek
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000, Ljubljana, Slovenia
| | - Tjaša Goričan
- Laboratory for Molecular Structural Dynamics, Theory Department, National Institute of Chemistry, Hajdrihova 19, 1001, Ljubljana, Slovenia
| | - Marius Gedgaudas
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Vilnius University, Saulėtekio al. 7 (C319), LT-10257, Vilnius, Lithuania
| | - Živa Zajec
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000, Ljubljana, Slovenia
| | - Dunja Urbančič
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000, Ljubljana, Slovenia
| | - Ana Jug
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000, Ljubljana, Slovenia
| | - Žiga Skok
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000, Ljubljana, Slovenia
| | - Caterina Sturtzel
- St. Anna Children's Cancer Research Institute, Zimmermannplatz 10, 1090, Vienna, Austria
| | - Martin Distel
- St. Anna Children's Cancer Research Institute, Zimmermannplatz 10, 1090, Vienna, Austria
| | - Simona Golič Grdadolnik
- Laboratory for Molecular Structural Dynamics, Theory Department, National Institute of Chemistry, Hajdrihova 19, 1001, Ljubljana, Slovenia
| | - Kesavan Babu
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105-3678, USA
| | - Ashna Panchamatia
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105-3678, USA
| | - Timothy R Stachowski
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105-3678, USA
| | - Marcus Fischer
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105-3678, USA
| | - Janez Ilaš
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000, Ljubljana, Slovenia
| | - Asta Zubrienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Vilnius University, Saulėtekio al. 7 (C319), LT-10257, Vilnius, Lithuania
| | - Daumantas Matulis
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Vilnius University, Saulėtekio al. 7 (C319), LT-10257, Vilnius, Lithuania
| | - Nace Zidar
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000, Ljubljana, Slovenia.
| | - Tihomir Tomašič
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000, Ljubljana, Slovenia.
| |
Collapse
|
37
|
Qu X, Wang S, Zhao S, Wan C, Xu W, Huang C. The dynamic triage interplay of Hsp90 with its chaperone cycle and client binding. Nat Commun 2024; 15:10661. [PMID: 39663352 PMCID: PMC11634960 DOI: 10.1038/s41467-024-55026-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/28/2024] [Indexed: 12/13/2024] Open
Abstract
Hsp90, a crucial molecular chaperone, regulates diverse client proteins, impacting both normal biology and disease. Central to its function is its conformational plasticity, driven by ATPase activity and client interactions. However, comprehensive insights into Hsp90's dynamic molecular transitions remain elusive. Using solution NMR spectroscopy, we reveal how ATP binding, hydrolysis, and client engagement drive conformational and dynamic shifts in E. coli Hsp90, HtpG, through its chaperone cycle. Pronounced conformational fluctuations occur, especially in regions crucial for nucleotide binding and conformational transitions. ATP binding induces slow-exchanging conformations, representing discrete on-path transition states from open to closed forms, while ATP hydrolysis shifts HtpG into a compact conformation. Client binding acts as an allosteric switch, dynamically priming HtpG for elevated chaperone activity and, therefore, its efficient remodeling. Here, we provide atomic-level insights into Hsp90's functional mechanism, highlighting the interplay of conformation, dynamics, nucleotide, and client interactions.
Collapse
Affiliation(s)
- Xiaozhan Qu
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Laboratory for Physical Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P.R. China
- Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou, 450001, China
| | - Simin Wang
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Laboratory for Physical Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P.R. China
| | - Shuo Zhao
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Laboratory for Physical Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P.R. China
| | - Chanjuan Wan
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Laboratory for Physical Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P.R. China
| | - Weiya Xu
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Laboratory for Physical Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P.R. China.
| | - Chengdong Huang
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Laboratory for Physical Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P.R. China.
| |
Collapse
|
38
|
Aguilar-Rodríguez J, Jakobson CM, Jarosz DF. The Hsp90 Molecular Chaperone as a Global Modifier of the Genotype-Phenotype-Fitness Map: An Evolutionary Perspective. J Mol Biol 2024; 436:168846. [PMID: 39481633 PMCID: PMC11608137 DOI: 10.1016/j.jmb.2024.168846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/02/2024]
Abstract
Global modifier genes influence the mapping of genotypes onto phenotypes and fitness through their epistatic interactions with genetic variants on a massive scale. The first such factor to be identified, Hsp90, is a highly conserved molecular chaperone that plays a central role in protein homeostasis. Hsp90 is a "hub of hubs" that chaperones proteins engaged in many key cellular and developmental regulatory networks. These clients, which are enriched in kinases, transcription factors, and E3 ubiquitin ligases, drive diverse cellular functions and are themselves highly connected. By contrast to many other hub proteins, the abundance and activity of Hsp90 changes substantially in response to shifting environmental conditions. As a result, Hsp90 modifies the functional impact of many genetic variants simultaneously in a manner that depends on environmental stress. Studies in diverse organisms suggest that this coupling between Hsp90 function and challenging environments exerts a substantial impact on what parts of the genome are visible to natural selection, expanding adaptive opportunities when most needed. In this Perspective, we explore the multifaceted role of Hsp90 as global modifier of the genotype-phenotype-fitness map as well as its implications for evolution in nature and the clinic.
Collapse
Affiliation(s)
- José Aguilar-Rodríguez
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA; Department of Biology, Stanford University, Stanford, CA, USA
| | - Christopher M Jakobson
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel F Jarosz
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA; Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
39
|
Wang W, Zhai S, Yang W, Gao H, Chang N, Zhang M, Hou Y, Bai G. Acacetin alleviates rheumatoid arthritis by targeting HSP90 ATPase domain to promote COX-2 degradation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156171. [PMID: 39489991 DOI: 10.1016/j.phymed.2024.156171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 10/01/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Inflammation plays a significant role in initiating and sustaining rheumatoid arthritis (RA). Acacetin, a natural flavonoid compound, exhibits excellent anti-inflammatory effects specifically for RA. However, its relevant targets and molecular mechanisms remain to be elucidated. PURPOSE This study aims to investigate the mechanism of acacetin in the therapeutic efficacy of acacetin in RA and search for new therapeutic options for RA treatment. METHODS A collagen-induced RA mouse model was established to evaluate the therapeutic effect of acacetin. Acacetin functional probes were synthesized to capture potential target proteins in RAW264.7 cells. Various small molecule-protein interaction methods were conducted to verify the binding of acacetin to target protein. Molecular docking and site directed mutagenesis tests were performed to analyze the specific binding sites. Co-immunoprecipitation, immunofluorescence assay and western blot were engineered to explore the effect of acacetin on COX-2 degradation by targeting HSP90. RESULTS Acacetin specifically binds to the ATP domain of HSP90, to facilitate the dissociation between HSP90 and COX-2, inducing the ubiquitin-degradation of COX-2 in macrophages. Acacetin suppressed the production of pro-inflammatory cytokines, as well as inflammatory related pathways, exerting excellent anti-inflammatory effects in RA. CONCLUSIONS This research proved that acacetin, a novel HSP90 ATPase inhibitor, inhibits the functional folding of the client protein COX-2, promoting its ubiquitin degradation for anti-inflammation. Targeting HSP90 is a viable strategy to inhibit inflammation, affording a distinct way to managing joint inflammation and pains associated with RA.
Collapse
Affiliation(s)
- Wenshuang Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, China
| | - Shanshan Zhai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, China
| | - Wen Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, China
| | - He Gao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, China
| | - Nianwei Chang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Man Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, China
| | - Yuanyuan Hou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, China.
| | - Gang Bai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, China
| |
Collapse
|
40
|
Fulton MD, Yama DJ, Dahl E, Johnson JL. Hsp90 and cochaperones have two genetically distinct roles in regulating eEF2 function. PLoS Genet 2024; 20:e1011508. [PMID: 39652595 DOI: 10.1371/journal.pgen.1011508] [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: 04/12/2024] [Revised: 12/17/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Protein homeostasis relies on the accurate translation and folding of newly synthesized proteins. Eukaryotic elongation factor 2 (eEF2) promotes GTP-dependent translocation of the ribosome during translation. eEF2 folding was recently shown to be dependent on Hsp90 as well as the cochaperones Hgh1, Cns1, and Cpr7. We examined the requirement for Hsp90 and cochaperones more closely and found that Hsp90 and cochaperones have two distinct roles in regulating eEF2 function. Yeast expressing one group of Hsp90 mutations or one group of cochaperone mutations had reduced steady-state levels of eEF2. The growth of Hsp90 mutants that affected eEF2 accumulation was also negatively affected by deletion of the gene encoding Hgh1. Further, mutations in yeast eEF2 that mimic disease-associated mutations in human eEF2 were negatively impacted by loss of Hgh1 and growth of one mutant was partially rescued by overexpression of Hgh1. In contrast, yeast expressing different groups of Hsp90 mutations or a different cochaperone mutation had altered sensitivity to diphtheria toxin, which is dictated by a unique posttranslational modification on eEF2. Our results provide further evidence that Hsp90 contributes to proteostasis not just by assisting protein folding, but also by enabling accurate translation of newly synthesized proteins. In addition, these results provide further evidence that yeast Hsp90 mutants have distinct in vivo effects that correlate with defects in subsets of cochaperones.
Collapse
Affiliation(s)
- Melody D Fulton
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Danielle J Yama
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Ella Dahl
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Jill L Johnson
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| |
Collapse
|
41
|
Li J, Qiu H, Dong Q, Yu H, Piao C, Li Z, Sun Y, Cui X. Androgen-targeted hsa_circ_0085121 encodes a novel protein and improves the development of prostate cancer through facilitating the activity of PI3K/Akt/mTOR pathway and enhancing AR-V7 alternative splicing. Cell Death Dis 2024; 15:848. [PMID: 39567496 PMCID: PMC11579034 DOI: 10.1038/s41419-024-07246-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024]
Abstract
Prostate cancer (PCa) is the most prevalent type of cancer and the second leading cause of mortality in males, with a marked increase in incidence observed across the globe. In the present study, whole-transcriptome analysis was conducted to identify differentially expressed circular RNAs (DE-circRNAs). The coding abilities of the DE-circRNAs were analyses, and it was found that hsa_circ_0085121 (circRNF19A) not only exhibited overexpression in PCa cells and tumor samples, but also encoded a 490 amino acid polypeptide designated circRNF19A-490aa. The knockdown of circRNF19A was observed to notably inhibit the proliferation, invasion, migration and docetaxel resistance of PCa cells. In contrast, mutation of the IRES significantly impaired the tumor-promoting function of circRNF19A, indicating that circRNF19A-490aa is the primary form that regulates the malignant behaviors of PCa cells. Mechanistically, circRNF19A-490aa was demonstrated to interact with HSP90AA1, thereby enhancing AR activity and facilitating the activation of the Akt/mTOR and PLK1 pathways. Furthermore, circRNF19A-490aa was observed to interact with HNRNPF, facilitating the recruitment of HNRNPF to the splicing site of AR-V7 and enhancing its alternative splicing. Finally, the androgen receptor (AR) was observed to bind to the promoter region of the RNF19A gene, subsequently regulating the expression of circRNF19A and circRNF19A-490aa. These data indicate that circRNF19A plays a pivotal role in AR activation and AR-V7 generation by encoding a novel protein, circRNF19A-490aa, and targeting circRNF19A may prove an effective strategy for impeding the progression of CRPC.
Collapse
Affiliation(s)
- Jianfeng Li
- Department of Urology, First Hospital of China Medical University, #155 Nanjing North Road, 110001, Shenyang, China
| | - Hui Qiu
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, #36 Sanhao Street, 110004, Shenyang, China
| | - Qingzhuo Dong
- Department of Urology, First Hospital of China Medical University, #155 Nanjing North Road, 110001, Shenyang, China
| | - Hongyuan Yu
- Department of Urology, First Hospital of China Medical University, #155 Nanjing North Road, 110001, Shenyang, China
| | - Chiyuan Piao
- Department of Urology, First Hospital of China Medical University, #155 Nanjing North Road, 110001, Shenyang, China
| | - Zhengxiu Li
- Department of Dermatology, First Hospital of China Medical University, #155 Nanjing North Road, 110001, Shenyang, China
| | - Yanbin Sun
- Department of Thoracic Surgery, First Hospital of China Medical University, #155 Nanjing North Road, 110001, Shenyang, China.
| | - Xiaolu Cui
- Department of Urology, First Hospital of China Medical University, #155 Nanjing North Road, 110001, Shenyang, China.
| |
Collapse
|
42
|
Keshavarzipour F, Abbasi M, Khorsandi Z, Ardestani M, Sadeghi-Aliabadi H. Design, synthesis and biological studies of new isoxazole compounds as potent Hsp90 inhibitors. Sci Rep 2024; 14:28017. [PMID: 39543364 PMCID: PMC11564562 DOI: 10.1038/s41598-024-79051-5] [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] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
Heat shock protein 90 (Hsp90), a molecular chaperone, contributes to the preservation of folding, structure, stability, and function proteins. In this study, novel compounds comprising isoxazole structure were designed, synthesized and their potential ability as Hsp90 inhibitors was validated through docking studies. The active site-based compounds were prepared through a multi-step synthesis process and their chemical structures were characterized employing FT-IR, NMR, and mass spectrometry analysis. Cytotoxic and Hsp90 inhibition activities of synthesized compounds were assessed by MTT assay and ELISA kit, respectively. Based on the obtained results, compound 5 exhibited the highest cytotoxicity (IC50; 14 µM) against cancer cells and reduced Hsp90 expression from 5.54 ng/mL in untreated (normal cells) to 1.56 ng/mL in cancer cells. Moreover, molecular dynamics (MD) simulation results indicated its high affinity to target protein and approved its excellent stability which is essential for exerting an inhibitory effect on cancer cell proliferation.
Collapse
Affiliation(s)
- Fariba Keshavarzipour
- Pharmaceutical Sciences Research Centre, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, 81746-7346, Isfahan, Iran
| | - Maryam Abbasi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Hormozgan University of Medical Sciences, 791969-1982, Bandar Abbas, Iran
| | - Zahra Khorsandi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Isfahan University of Medical Sciences, 81746- 7346, Isfahan, Iran
| | - Mina Ardestani
- Department of Medicinal Chemistry, Faculty of Pharmacy, Isfahan University of Medical Sciences, 81746- 7346, Isfahan, Iran
| | - Hojjat Sadeghi-Aliabadi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Isfahan University of Medical Sciences, 81746- 7346, Isfahan, Iran.
| |
Collapse
|
43
|
Reynolds TS, Blagg BSJ. Extracellular heat shock protein 90 alpha (eHsp90α)'s role in cancer progression and the development of therapeutic strategies. Eur J Med Chem 2024; 277:116736. [PMID: 39126794 PMCID: PMC11374465 DOI: 10.1016/j.ejmech.2024.116736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024]
Abstract
Heat shock protein 90 alpha (Hsp90α) is an abundantly expressed and evolutionarily conserved molecular chaperone. Hsp90α is the inducible Hsp90 isoform, and its expression and secretion extracellularly (eHsp90α) can be triggered in response to a variety of cellular stresses to protect/activate client proteins and to facilitate cellular adjustment to the stress. As a result, cancers often have high expression levels of intracellular and extracellular (plasma) Hsp90α, allowing them to support their oncogenesis and progression. In fact, (e)Hsp90α has been implicated in regulating processes such as cell signaling transduction, DNA repair, promotion of the Epithelial-to-Mesenchymal Transition (EMT), promotion of angiogenesis, immune response, and cell migration. Hsp90α levels have been correlated with cancer progression and severity in several cancers, indicating that it may be a useful biomarker or drug-target for cancer. To date, the development of intracellular Hsp90α-targeted therapies include standard N-terminal ATP-competitive inhibitors and allosteric regulators that bind to Hsp90α's middle or C-terminal domain. On-target toxicities and dosing complications as a result of Hsp90α inhibition has driven the development of eHsp90α-targeted therapies. Examples include anti-Hsp90α monoclonal antibodies and cell-impermeable Hsp90α small molecule inhibitors. This review aims to discuss the many roles Hsp90α plays in cancer progression with a focus on the current development of Hsp90α-targeted therapies.
Collapse
Affiliation(s)
- Tyelor S Reynolds
- Department of Chemistry and Biochemistry, The University of Notre Dame, 305 McCourtney Hall, Notre Dame, IN, 46556, USA
| | - Brian S J Blagg
- Department of Chemistry and Biochemistry, The University of Notre Dame, 305 McCourtney Hall, Notre Dame, IN, 46556, USA.
| |
Collapse
|
44
|
Chen L, Guo X, Lin W, Huang Y, Zhuang S, Li Q, Xu J, Ye S. Curcumin derivative C210 induces Epstein-Barr virus lytic cycle and inhibits virion production by disrupting Hsp90 function. Sci Rep 2024; 14:26694. [PMID: 39496752 PMCID: PMC11535535 DOI: 10.1038/s41598-024-77294-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/21/2024] [Indexed: 11/06/2024] Open
Abstract
Lytic induction therapy was devised to selectively combat malignancies associated with Epstein-Barr virus (EBV) by triggering viral reactivation from latency. At present, the major challenges of lytic induction therapy are to maximize reactivating efficiencies and meanwhile minimize infectious virion production. C210, a novel curcumin derivative with potent Hsp90 inhibitory activity, was explored for EBV-reactivating and virion-producing effects in EBV-positive nasopharyngeal carcinoma (NPC) and gastric carcinoma (GC) cell lines. And the molecular mechanisms underlying these effects were determined. Follow C210 treatment, EBV lytic RNAs and proteins were upregulated, but infectious virions were not produced. Knockdown of heat shock protein 90 (Hsp90) induced expression of lytic RNAs and proteins, and diminished C210-driven EBV lytic induction. Pretreatment with an X box binding protein 1 (XBP1) inhibitor reduced C210-induced EBV lytic RNA. Furthermore, we demonstrated that C210 inhibited the binding of Hsp90 with its clients, signal transducer and activator of transcription 3 (STAT3) and xeroderma pigmentosum group B-complementing protein (XPB), which subsequently promoted their proteasomal degradation. Degradation of STAT3 by C210 enhanced the EBV-reactivating and anticancer capacity of suberoylanilide hydroxamic acid (SAHA). Depletion of XPB blocked SAHA-induced expression of late viral genes and production of infectious virions. These results elucidate a novel Hsp90 inhibitor targeting EBV lytic phase and extend the research on lytic induction strategy, which may offer reference value in the treatment of EBV-positive malignancies.
Collapse
Grants
- 2019Y9131 the Joint Funds for the Innovation of Science and Technology, Fujian province, China
- 2019Y9131 the Joint Funds for the Innovation of Science and Technology, Fujian province, China
- 2019Y9131 the Joint Funds for the Innovation of Science and Technology, Fujian province, China
- 2019Y9131 the Joint Funds for the Innovation of Science and Technology, Fujian province, China
- 2019Y9131 the Joint Funds for the Innovation of Science and Technology, Fujian province, China
- 2019Y9131 the Joint Funds for the Innovation of Science and Technology, Fujian province, China
- 2019Y9131 the Joint Funds for the Innovation of Science and Technology, Fujian province, China
- 2019Y9131 the Joint Funds for the Innovation of Science and Technology, Fujian province, China
- 2022QH2038 the Startup Fund for scientific research, Fujian Medical University
- 2022QH2038 the Startup Fund for scientific research, Fujian Medical University
- 2022QH2038 the Startup Fund for scientific research, Fujian Medical University
- 2022QH2038 the Startup Fund for scientific research, Fujian Medical University
- 2022QH2038 the Startup Fund for scientific research, Fujian Medical University
- 2022QH2038 the Startup Fund for scientific research, Fujian Medical University
- 2022QH2038 the Startup Fund for scientific research, Fujian Medical University
- 2022QH2038 the Startup Fund for scientific research, Fujian Medical University
Collapse
Affiliation(s)
- Linli Chen
- Department of Otorhinolaryngology Head and Neck Surgery, Fujian Institute of Otorhinolaryngology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
- Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiaojing Guo
- Department of Otorhinolaryngology Head and Neck Surgery, Fujian Institute of Otorhinolaryngology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
- Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Wen Lin
- Department of Otorhinolaryngology Head and Neck Surgery, Fujian Institute of Otorhinolaryngology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
- Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yingying Huang
- Department of Otorhinolaryngology Head and Neck Surgery, Fujian Institute of Otorhinolaryngology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
- Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Suling Zhuang
- Department of Otorhinolaryngology Head and Neck Surgery, Fujian Institute of Otorhinolaryngology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
- Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Qianfeng Li
- Department of Otorhinolaryngology Head and Neck Surgery, Fujian Institute of Otorhinolaryngology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
- Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jianhua Xu
- The School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, 350122, Fujian, China.
| | - Shengnan Ye
- Department of Otorhinolaryngology Head and Neck Surgery, Fujian Institute of Otorhinolaryngology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China.
- Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
45
|
Du J, Zhang J, Liu D, Gao L, Liao H, Chu L, Lin J, Li W, Meng X, Zou F, Cai S, Zou M, Dong H. 1G6-D7 Inhibits Homologous Recombination Repair by Targeting Extracellular HSP90α to Promote Apoptosis in Non-Small Cell Lung Cancer. ENVIRONMENTAL TOXICOLOGY 2024; 39:4884-4898. [PMID: 38899512 DOI: 10.1002/tox.24356] [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: 02/23/2024] [Revised: 05/07/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024]
Abstract
Despite recent advances in treatment, non-small cell lung cancer (NSCLC) continues to have a high mortality rate. Currently, NSCLC pathogenesis requires further investigation, and therapeutic drugs are still under development. Homologous recombination repair (HRR) repairs severe DNA double-strand breaks. Homologous recombination repair deficiency (HRD) occurs when HRR is impaired and causes irreparable double-strand DNA damage, leading to genomic instability and increasing the risk of cancer development. Poly(ADP-ribose) polymerase (PARP) inhibitors can effectively treat HRD-positive tumors. Extracellular heat shock protein 90α (eHSP90α) is highly expressed in hypoxic environments and inhibits apoptosis, thereby increasing cellular tolerance. Here, we investigated the relationship between eHSP90α and HRR in NSCLC. DNA damage models were established in NSCLC cell lines (A549 and H1299). The activation of DNA damage and HRR markers, apoptosis, proliferation, and migration were investigated. In vivo tumor models were established using BALB/c nude mice and A549 cells. We found that human recombinant HSP90α stimulation further activated HRR and reduced DNA damage extent; however, eHSP90α monoclonal antibody, 1G6-D7, effectively inhibited HRR. HRR inhibition and increased apoptosis were observed after LRP1 knockdown; this effect could not be reversed with hrHSP90α addition. The combined use of 1G6-D7 and olaparib caused significant apoptosis and HRR inhibition in vitro and demonstrated promising anti-tumor effects in vivo. Extracellular HSP90α may be involved in HRR in NSCLC through LRP1. The combined use of 1G6-D7 and PARP inhibitors may exert anti-tumor effects by inhibiting DNA repair and further inducing apoptosis of NSCLC cells.
Collapse
Affiliation(s)
- Jiangzhou Du
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinming Zhang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dongyu Liu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lin Gao
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hua Liao
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lanhe Chu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jie Lin
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wei Li
- Department of Dermatology, The USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, California, Los Angeles, USA
| | - Xiaojing Meng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Fei Zou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Shaoxi Cai
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mengchen Zou
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hangming Dong
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
46
|
An J, Park H, Ju M, Woo Y, Seo Y, Min J, Lee T. An updated review on the development of a nanomaterial-based field-effect transistor-type biosensors to detect exosomes for cancer diagnosis. Talanta 2024; 279:126604. [PMID: 39068827 DOI: 10.1016/j.talanta.2024.126604] [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: 03/29/2024] [Revised: 06/24/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Cancer, a life-threatening genetic disease caused by abnormalities in normal cell growth regulatory functions, poses a significant challenge that current medical technologies cannot fully overcome. The current desired breakthrough is to diagnose cancer as early as possible and increase survival rates through treatments tailored to the prognosis and appropriate follow-up. From a perspective that reflects this contemporary paradigm of cancer diagnostics, exosomes are emerging as promising biomarkers. Exosomes, serving as mobile biological information repositories of cancer cells, have been known to create a microtumor environment in surrounding cells, and significant insight into the clinical significance of cancer diagnosis targeting them has been reported. Therefore, there are growing interests in constructing a system that enables continuous screening with a focus on patient-friendly and flexible diagnosis, aiming to improve cancer screening rates through exosome detection. This review focuses on a proposed exosome-embedded biological information-detecting platform employing a field-effect transistor (FET)-based biosensor that leverages portability, cost-effectiveness, and rapidity to minimize the stages of sacrifice attributable to cancer. The FET-applied biosensing technique, stemming from variations in an electric field, is considered an early detection system, offering high sensitivity and a prompt response frequency for the qualitative and quantitative analysis of biomolecules. Hence, an in-depth discussion was conducted on the understanding of various exosome-based cancer biomarkers and the clinical significance of recent studies on FET-based biosensors applying them.
Collapse
Affiliation(s)
- Jeongyun An
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-Ro, Nowon-Gu, Seoul, 01897, Republic of Korea
| | - Hyunjun Park
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-Ro, Nowon-Gu, Seoul, 01897, Republic of Korea
| | - Minyoung Ju
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-Ro, Nowon-Gu, Seoul, 01897, Republic of Korea
| | - Yeeun Woo
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-Ro, Nowon-Gu, Seoul, 01897, Republic of Korea
| | - Yoshep Seo
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-Ro, Nowon-Gu, Seoul, 01897, Republic of Korea
| | - Junhong Min
- School of Integrative Engineering, Chung-Ang University, Dongjak-Gu, Seoul, 06974, Republic of Korea.
| | - Taek Lee
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-Ro, Nowon-Gu, Seoul, 01897, Republic of Korea.
| |
Collapse
|
47
|
Naoki K, Igawa S, Uojima H, Tsumura H, Sengoku N, Karayama M, Shimomura A, Ohtake T, Shio Y, Hosokawa A, Komatsu Y, Kumagai Y. Cardiovascular safety of pimitespib in patients with advanced solid tumors: An open-label, nonrandomized, phase 1 study. Cancer 2024; 130:3745-3756. [PMID: 38985885 DOI: 10.1002/cncr.35447] [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: 02/02/2024] [Revised: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Pimitespib (TAS-116), a first-in-class, oral, selective heat-shock protein 90 inhibitor, is approved as fourth-line treatment for gastrointestinal stromal tumors in Japan. This phase 1 study evaluated the cardiac safety of pimitespib. METHODS In this open-label, nonrandomized, multicenter study, Japanese patients (aged ≥20 years) with refractory, advanced solid tumors received placebo on day -1, then pimitespib 160 mg daily on days 1-5 of the cardiac safety evaluation period. Electrocardiograms were conducted at baseline, and on days -2, -1, 1, and 5; and blood samples were collected on days 1 and 5. Patients then received once-daily pimitespib for 5 days every 3 weeks. The primary end point was the time-matched difference in QT interval corrected for heart rate using the Fridericia correction (QTcF) between pimitespib and placebo. Pharmacokinetics, safety, and preliminary efficacy were also assessed. RESULTS Of the 22 patients in the cardiac safety-evaluable population, no clinically relevant QTc prolongation was observed; the upper bound of the one-sided 95% confidence interval for the time-matched difference in change from baseline in QTcF was <20 msec at all time points on days 1 and 5. Pimitespib pharmacokinetic parameters were consistent with previous data, and the time-matched difference in change from baseline in QTcF showed no marked increase as plasma concentrations increased. The safety profile was acceptable; 40% of patients experienced grade 3 or greater adverse drug reactions, mostly diarrhea (20%). The median progression-free survival was 3.1 months. CONCLUSIONS In Japanese patients with refractory, advanced solid tumors, pimitespib was not associated with clinically relevant QTc prolongation, and there were no cardiovascular safety concerns. PLAIN LANGUAGE SUMMARY Pimitespib is a new anticancer drug that is being used to treat cancer in the stomach or intestines (gastrointestinal stromal tumors). This study demonstrated that pimitespib had no marked effect on heart rhythm or negative effects on the heart or blood vessels and had promising anticancer effects in Japanese patients with advanced solid tumors who were unable to tolerate or benefit from standard treatment.
Collapse
Affiliation(s)
- Katsuhiko Naoki
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Satoshi Igawa
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Haruki Uojima
- Department of Gastroenterology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Hideyasu Tsumura
- Department of Urology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Norihiko Sengoku
- Department of Breast and Endocrine Surgery, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Masato Karayama
- Department of Chemotherapy, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Akihiko Shimomura
- Department of Breast and Medical Oncology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tohru Ohtake
- Department of Breast Surgery, Fukushima Medical University Hospital, Fukushima, Japan
| | - Yutaka Shio
- Department of Chest Surgery, Fukushima Medical University Hospital, Fukushima, Japan
| | - Ayumu Hosokawa
- Department of Clinical Oncology, University of Miyazaki Hospital, Miyazaki, Japan
| | - Yoshito Komatsu
- Department of Cancer Center, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Yuji Kumagai
- Department of Research, Clinical Trial Center, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
| |
Collapse
|
48
|
Huang M, Chen L, Ma X, Xu H. Celastrol attenuates the invasion and migration and augments the anticancer effects of olaparib in prostate cancer. Cancer Cell Int 2024; 24:352. [PMID: 39462410 PMCID: PMC11514812 DOI: 10.1186/s12935-024-03542-8] [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: 07/25/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) is a leading malignancy among men globally, with rising incidence rates emphasizing the critical need for better detection and therapeutic approaches. The roles of HSP90AB1 and PARP1 in prostate cancer cells suggest potential targets for enhancing treatment efficacy. METHODS This study investigated the overexpression of HSP90AB1 and PARP1 in prostate cancer cells and the impact of HSP90AB1 knockdown on the sensitivity of these cells to the PARP inhibitor olaparib. We also explored the combined effect of olaparib and celastrol, an HSP90 inhibitor, on the clonogenic survival, migration, proliferation, and overall viability of prostate cancer cells, alongside the modulation of the PI3K/AKT pathway. An in vivo PC3 xenograft mouse model was used to assess the antitumor effects of the combined treatment. RESULTS Our findings revealed significant overexpression of HSP90AB1 and PARP1 in prostate cancer cells. Knockdown of HSP90AB1 increased cell sensitivity to olaparib. The combination of olaparib and celastrol significantly reduced prostate cancer cell survival, migration, proliferation, and enhanced cumulative DNA damage. Celastrol also downregulated the PI3K/AKT pathway, increasing cell susceptibility to olaparib. In vivo experiments demonstrated that celastrol and olaparib together exerted strong antitumor effects. CONCLUSIONS The study indicates that targeting both HSP90AB1 and PARP1 presents a promising therapeutic strategy for prostate cancer. The synergistic combination of celastrol and olaparib enhances the efficacy of treatment against prostate cancer, offering a potent approach to combat this disease.
Collapse
Affiliation(s)
- Mengqiu Huang
- College of Biology and Environmental Engineering, Guiyang University, Guiyang, Guizhou, China, 550005
| | - Lin Chen
- Department of Ophthalmology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiaoyan Ma
- College of Food and Pharmaceutical Engineering, Guizhou Institute of Technology, Guiyang, Guizhou, China
| | - Houqiang Xu
- College of Biology and Environmental Engineering, Guiyang University, Guiyang, Guizhou, China, 550005.
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, Guizhou, China.
- College of Animal Science, Guizhou University, Guiyang, Guizhou, China.
- Department of Biomedicine, Guizhou University school of Medicine, 2708#, Huaxi Road South, Huaxi District, Guiyang, 550025, Guizhou, China.
| |
Collapse
|
49
|
Kitson RRA, Kitsonová D, Siegel D, Ross D, Moody CJ. Geldanamycin, a Naturally Occurring Inhibitor of Hsp90 and a Lead Compound for Medicinal Chemistry. J Med Chem 2024; 67:17946-17963. [PMID: 39361055 PMCID: PMC11513894 DOI: 10.1021/acs.jmedchem.4c01048] [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: 05/03/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 10/25/2024]
Abstract
Geldanamycin remains a driver in the medicinal chemistry of heat shock protein 90 (Hsp90) inhibition, even half a century after its original isolation from nature. This Perspective focuses on the properties of the benzoquinone ring of the natural product that enable a range of functionalization reactions to take place. Therefore, inherent reactivity at C-17, where the methoxy group serves as a vinylogous ester, and at C-19 that demonstrates nucleophilic, enamide-type character toward electrophiles, and also as a conjugate acceptor to react with nucleophiles, has facilitated the synthesis of semisynthetic derivatives. Thus, a range of C-17-substituted amine derivatives has been investigated in oncology applications, with a number of compounds in this series reaching clinical trials. In contrast, the 19-position of geldanamycin has received less attention, although 19-substituted derivatives offer promise with markedly reduced toxicity compared to geldanamycin itself, while retaining Hsp90 inhibitory activity albeit with diminished potency in cellular studies.
Collapse
Affiliation(s)
- Russell R. A. Kitson
- Department
of Organic and Bioorganic Chemistry, Charles
University, Faculty of Pharmacy in Hradec Králové, Akademika Heyrovského 1203, 50005 Hradec Králové, Czech Republic
| | - Dominika Kitsonová
- Datwyler
Sealing Technologies CZ Ltd., Polní 224, 50401 Nový Bydžov, Czech
Republic
| | - David Siegel
- Department
of Pharmaceutical Sciences, University of
Colorado Anschutz Medical Campus, 12850 East Montview Boulevard, Aurora, Colorado 80045, United States
| | - David Ross
- Department
of Pharmaceutical Sciences, University of
Colorado Anschutz Medical Campus, 12850 East Montview Boulevard, Aurora, Colorado 80045, United States
| | - Christopher J. Moody
- School
of Chemistry, University of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| |
Collapse
|
50
|
Stewart M, Schisler JC. Targeting chaperone modifications: Innovative approaches to cancer treatment. J Biol Chem 2024; 300:107907. [PMID: 39433125 PMCID: PMC11599458 DOI: 10.1016/j.jbc.2024.107907] [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: 05/28/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024] Open
Abstract
Cancer and other chronic diseases are marked by alterations in the protein quality control system, affecting the posttranslational destiny of various proteins that regulate, structure, and catalyze cellular processes. Cellular chaperones, also known as heat shock proteins (HSPs), are pivotal in this system, performing protein triage that often determines the fate of proteins they bind to. Grasping the regulatory mechanisms of HSPs and their associated cofactors is crucial for understanding protein quality control in both healthy and diseased states. Recent research has shed light on the interactions within the protein quality control system and how post-translational modification govern protein interactions, function, and localization, which can drive or inhibit cell proliferation. This body of work encompasses critical elements of the heat shock response, including heat shock protein 70, heat shock protein 90, carboxyl-terminus of HSC70 interacting protein, and heat shock protein organizing protein. This review aims to synthesize these advancements, offering a holistic understanding of the system and its response when commandeered by diseases like cancer. We focus on the mechanistic shift in co-chaperone engagement-transitioning from heat shock protein organizing protein to carboxyl-terminus of HSC70 interacting protein in association with heat shock protein 70 and heat shock protein 90-which could influence cellular growth and survival pathways. A comprehensive examination of posttranslational modification-driven regulation within the protein quality control network is presented, highlighting the roles of activation factors, chaperones, and co-chaperones. Our insights aim to inform new strategies for therapeutically targeting diseases by considering the entire heat shock response system.
Collapse
Affiliation(s)
- Mariah Stewart
- The McAllister Heart Institute and Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jonathan C Schisler
- The McAllister Heart Institute and Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; The Department of Pathology and Lab Medicine and Computational Medicine Program, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
| |
Collapse
|