1
|
Toulotte F, Coevoet M, Liberelle M, Bailly F, Zagiel B, Gelin M, Allemand F, Fourquet P, Melnyk P, Guichou JF, Cotelle P. Towards the design of ligands of the internal pocket TEADs C-terminal domain. Eur J Med Chem 2025; 282:117026. [PMID: 39571457 DOI: 10.1016/j.ejmech.2024.117026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/17/2024] [Accepted: 11/01/2024] [Indexed: 12/10/2024]
Abstract
The Hippo pathway controls in organ size and tissue homeostasis through regulating cell growth, proliferation and apoptosis. Phosphorylation of the transcription co-activator YAP (Yes associated protein) and TAZ (Transcriptional coactivator with PDZ-binding motif) regulates their nuclear import and therefore their interaction with TEAD (Transcriptional Enhanced Associated Domain). YAP, TAZ and TEADs are dysregulated in several solid cancers making YAP/TAZ-TEAD interaction a new anti-cancer target. We identified by screening a small in-house library, 5-benzyloxindole which binds to hTEAD2 at its internal/palmitate pocket. Its optimization led to covalent inhibitors bearing different warhead. Soaking with hTEAD2 gave seven new crystal structures where the ligands occupied palmitate pocket. 5-Benzyloxyindoles armed with vinylsulfamide moiety inhibit YAP/TAZ-TEAD target genes expression and breast cancer cell proliferation at micromolar concentration.
Collapse
Affiliation(s)
- Florine Toulotte
- Univ Lille, INSERM, CHU Lille, UMR-S 1172, Lille Neuroscience and Cognition Research Center, F-59000, Lille, France
| | - Mathilde Coevoet
- Univ Lille, INSERM, CHU Lille, UMR-S 1172, Lille Neuroscience and Cognition Research Center, F-59000, Lille, France
| | - Maxime Liberelle
- Univ Lille, INSERM, CHU Lille, UMR-S 1172, Lille Neuroscience and Cognition Research Center, F-59000, Lille, France
| | - Fabrice Bailly
- Univ Lille, INSERM, CHU Lille, UMR-S 1172, Lille Neuroscience and Cognition Research Center, F-59000, Lille, France
| | - Benjamin Zagiel
- Univ Lille, INSERM, CHU Lille, UMR-S 1172, Lille Neuroscience and Cognition Research Center, F-59000, Lille, France
| | - Muriel Gelin
- Centre de Biologie Structurale (CBS), CNRS, INSERM, Univ Montpellier, F-34090, Montpellier, France
| | - Frédéric Allemand
- Centre de Biologie Structurale (CBS), CNRS, INSERM, Univ Montpellier, F-34090, Montpellier, France
| | - Patrick Fourquet
- Centre de Recherche en Cancérologie de Marseille (CRCM), CNRS, INSERM, Marseille Protéomique, Institut Paoli-Calmettes, Aix Marseille University, 27 Bvd Leï Roure, CS 30059, 13273 Marseille, France
| | - Patricia Melnyk
- Univ Lille, INSERM, CHU Lille, UMR-S 1172, Lille Neuroscience and Cognition Research Center, F-59000, Lille, France.
| | - Jean-François Guichou
- Centre de Biologie Structurale (CBS), CNRS, INSERM, Univ Montpellier, F-34090, Montpellier, France.
| | - Philippe Cotelle
- Univ Lille, INSERM, CHU Lille, UMR-S 1172, Lille Neuroscience and Cognition Research Center, F-59000, Lille, France; ENSCL-Centrale Lille, CS 90108, F-59652, Villeneuve d'Ascq, France
| |
Collapse
|
2
|
Heinrich T, Schwarz D, Petersson C, Gunera J, Garg S, Schneider R, Keil M, Grimmeisen L, Unzue Lopez A, Albers L, Schlesiger S, Gambardella A, Bomke J, Carswell E, Schilke H, Diehl P, Doerfel B, Musil D, Trivier E, Broome R, Marshall S, Balsiger A, Friedrich E, Lemos AR, Santos SP, Sousa PMF, Freire F, Bandeiras TM, Bortoluzzi A, Wienke D. MoA Studies of the TEAD P-Site Binding Ligand MSC-4106 and Its Optimization to TEAD1-Selective Amide M3686. J Med Chem 2024. [PMID: 39704449 DOI: 10.1021/acs.jmedchem.4c01949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Taking the structural information into account, we were able to tune the TEAD selectivity for a specific chemotype. However, different TEAD selectivity profiles did not affect the compound potency or efficacy in the NCI-H226 viability assay. Amides based on MSC-4106 or analogues showed improved viability efficacy compared with the corresponding acids. The amide M3686 exhibited AUC-driven efficacy in NCI-H226 xenograft models and had an improved 25-fold lower human dose prediction than MSC-4106. MSC-4106 was also used in HDX-MS studies to aid in the understanding of the MoA of P-site binding TEAD inhibitors. Artificial P-site binders rigidify certain areas in the periphery of the transcription factor that seem to be crucial for cofactor interaction, whereas a native fatty acid increased the protein dynamics of cofactor-binding interfaces.
Collapse
Affiliation(s)
- Timo Heinrich
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Daniel Schwarz
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Carl Petersson
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Jakub Gunera
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Sakshi Garg
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Richard Schneider
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Marina Keil
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Lisa Grimmeisen
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | | | - Lisa Albers
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Sarah Schlesiger
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | | | - Joerg Bomke
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Emma Carswell
- Cancer Research Horizons, Jonas Webb Building, Babraham Research Campus, CambridgeCB22 3AT, U.K
| | - Heike Schilke
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Patrizia Diehl
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Benjamin Doerfel
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Djordje Musil
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Elisabeth Trivier
- Cancer Research Horizons, 4NW, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, U.K
| | - Rebecca Broome
- Cancer Research Horizons, 4NW, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, U.K
| | - Sam Marshall
- Cancer Research Horizons, 4NW, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, U.K
| | - Alexander Balsiger
- Cancer Research Horizons, 4NW, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, U.K
| | - Erik Friedrich
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Ana R Lemos
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
| | - Sandra P Santos
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
| | - Pedro M F Sousa
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
| | - Filipe Freire
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
| | - Tiago M Bandeiras
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
| | - Alessio Bortoluzzi
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
| | - Dirk Wienke
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| |
Collapse
|
3
|
Carswell E, Heinrich T, Petersson C, Gunera J, Garg S, Schwarz D, Schlesiger S, Fischer F, Eichhorn T, Calder M, Smith G, MacDonald E, Wilson H, Hazel K, Trivier E, Broome R, Balsiger A, Sirohi S, Musil D, Freire F, Schilke H, Dillon C, Wienke D. Discovery of reversible and covalent TEAD 1 selective inhibitors MSC-1254 and MSC-5046 based on one scaffold. Bioorg Med Chem Lett 2024; 114:129981. [PMID: 39369801 DOI: 10.1016/j.bmcl.2024.129981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/06/2024] [Accepted: 09/27/2024] [Indexed: 10/08/2024]
Abstract
The Transcriptional Enhanced Associated Domain (TEAD) family of transcription factors are key components of the Hippo signalling family which play a crucial role in the regulation of cell proliferation, differentiation and apoptosis. The identification of inhibitors of the TEAD transcription factors are an attractive strategy for the development of novel anticancer therapies. A HTS campaign identified hit 1, which was optimised using structure-based drug design, to deliver potent TEAD1 selective inhibitors with both a reversible and covalent mode of inhibition. The preference for TEAD1 could be rationalised by steric differences observed in the lower pocket of the palmitoylation-site between subtypes, with TEAD1 having the largest available volume to accommodate substitution in this region.
Collapse
Affiliation(s)
- Emma Carswell
- Cancer Research Horizons, Jonas Webb Building, Babraham Research Campus, Cambridge CB22 3AT, UK.
| | - Timo Heinrich
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Carl Petersson
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Jakub Gunera
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Sakshi Garg
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Daniel Schwarz
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Sarah Schlesiger
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Frank Fischer
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Thomas Eichhorn
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Mathew Calder
- Cancer Research Horizons, Jonas Webb Building, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Geoffrey Smith
- Cancer Research Horizons, Jonas Webb Building, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Ellen MacDonald
- Cancer Research Horizons, Jonas Webb Building, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Hollie Wilson
- Cancer Research Horizons, Jonas Webb Building, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Katherine Hazel
- Cancer Research Horizons, Jonas Webb Building, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Elisabeth Trivier
- Cancer Research Horizons, 4NW, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Rebecca Broome
- Cancer Research Horizons, 4NW, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Alexander Balsiger
- Cancer Research Horizons, 4NW, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Sameer Sirohi
- Cancer Research Horizons, Jonas Webb Building, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Djordje Musil
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Filipe Freire
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
| | - Heike Schilke
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Christian Dillon
- Cancer Research Horizons, 4NW, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Dirk Wienke
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| |
Collapse
|
4
|
Koroleva OA, Kurkin AV, Shtil AA. The Hippo pathway as an antitumor target: time to focus on. Expert Opin Investig Drugs 2024; 33:1177-1185. [PMID: 39592955 DOI: 10.1080/13543784.2024.2432395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024]
Abstract
INTRODUCTION The Hippo signaling governs the expression of genes critically important for cell proliferation and survival. The components of this pathway are considered antitumor drug targets. However, the design of Hippo inhibitors is a challenge given the complexity of the network and redundancy of its elements. AREAS COVERED We review the current state-of-the-art in the structure of the Hippo pathway, the microenvironment-induced extracellular cues, the strategies to design pharmacological instruments for inactivation of the Hippo signaling using small molecular weight modulators, as well as the results of initial clinical trials. EXPERT OPINION One special characteristic of the Hippo signaling is the adverse role of phosphorylation: opposite to classical kinase cascades that activate the transcription factors, the Hippo kinases retain their partners in a transcriptionally inactive state. Therefore, approaches for pharmacological or genetic inhibition of Hippo protein kinases are counterproductive. The developing alternatives such as disruption of protein-protein interactions or PROTAC techniques are straightforward for preventing the Hippo signaling in cancer therapy.
Collapse
Affiliation(s)
- Olga A Koroleva
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Alexander V Kurkin
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Alexander A Shtil
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russian Federation
- Institute of Carcinogenesis, Blokhin National Medical Research Center of Oncology, Moscow, Russian Federation
| |
Collapse
|
5
|
Li Z, Li J, Cao Q, Shen T, Wang Y, He H, Tong M. Transcription factor TCF7L1 targeting HSPB6 is involved in EMT and PI3K/AKT/mTOR pathways in bladder cancer. J Biol Chem 2024; 301:108024. [PMID: 39608715 DOI: 10.1016/j.jbc.2024.108024] [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: 03/26/2024] [Revised: 11/06/2024] [Accepted: 11/17/2024] [Indexed: 11/30/2024] Open
Abstract
Bladder cancer is notorious for its high recurrence and costly treatment burden, prompting a search for novel therapeutic targets. Our study focuses on HSPB6, a small heat shock protein whose reduced expression in bladder cancer suggests a role in tumor biology. Using an integrative approach of bioinformatics, RNA sequencing, and cell-based assays, we show that HSPB6 upregulation inhibits cancer cell proliferation and metastasis while promoting apoptosis. Moreover, TCF7L1-mediated upregulation of HSPB6 leads to suppression of the PI3K/AKT/mTOR signaling pathway, a key driver of cancer progression. These results position HSPB6 as a compelling target for bladder cancer therapy, and its regulatory role in the PI3K/AKT/mTOR axis underscores its therapeutic potential. Our findings pave the way for future investigations into HSPB6-centered treatment strategies.
Collapse
Affiliation(s)
- Zizhi Li
- Department of Medicine, Soochow University, Soochow, Jiangsu, China; Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Junyi Li
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Qingfei Cao
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Tong Shen
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yingjie Wang
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Haoyang He
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Ming Tong
- Department of Medicine, Soochow University, Soochow, Jiangsu, China; Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China.
| |
Collapse
|
6
|
Paul S, Sims J, Pham T, Dey A. Targeting the Hippo pathway in cancer: kidney toxicity as a class effect of TEAD inhibitors? Trends Cancer 2024:S2405-8033(24)00223-1. [PMID: 39521692 DOI: 10.1016/j.trecan.2024.10.004] [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: 09/04/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024]
Abstract
The Hippo pathway has emerged as a critical player in both cancers and targeted therapy resistance. Recent drug discovery efforts have led to the development of TEAD inhibitors, several of which have already progressed to the clinic. To truly leverage their potential as anticancer therapeutics, safety considerations, particularly in regard to the kidney, warrant additional investigation. This review explores the Hippo pathway's role in cancers, its therapeutic potential, role in kidney development, and the need to evaluate the best strategies to translate its clinical application for long-term patient benefit.
Collapse
Affiliation(s)
- Sayantanee Paul
- Department of Discovery Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080
| | - Jessica Sims
- Department of Safety Assessment, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080
| | - Trang Pham
- Department of Discovery Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080
| | - Anwesha Dey
- Department of Discovery Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080.
| |
Collapse
|
7
|
Zhu R, Liu X, Zhang X, Zhong Z, Qi S, Jin R, Gu Y, Wang Y, Ling C, Chen K, Ye D, Yu FX. Gene therapy for diffuse pleural mesotheliomas in preclinical models by concurrent expression of NF2 and SuperHippo. Cell Rep Med 2024; 5:101763. [PMID: 39368484 PMCID: PMC11513813 DOI: 10.1016/j.xcrm.2024.101763] [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: 03/20/2024] [Revised: 08/03/2024] [Accepted: 09/12/2024] [Indexed: 10/07/2024]
Abstract
Diffuse pleural mesothelioma (DPM) is a lethal cancer with a poor prognosis and limited treatment options. The Hippo signaling pathway genes, such as NF2 and LATS1/2, are frequently mutated in DPM, indicating a tumor suppressor role in the development of DPM. Here, we show that in DPM cell lines lacking NF2 and in mice with a conditional Nf2 knockout, downregulation of WWC proteins, another family of Hippo pathway regulators, accelerates DPM progression. Conversely, the expression of SuperHippo, a WWC-derived minigene, effectively enhances Hippo signaling and suppresses DPM development. Moreover, the adeno-associated virus serotype 6 (AAV6) has been engineered to deliver both NF2 and SuperHippo genes into mesothelial cells, which substantially impedes tumor growth in xenograft and genetic DPM models and prolongs the median survival of mice. These findings serve as a proof of concept for the potential use of gene therapy targeting the Hippo pathway to treat DPM.
Collapse
Affiliation(s)
- Rui Zhu
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xincheng Liu
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xu Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhenxing Zhong
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Sixian Qi
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ruxin Jin
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yuan Gu
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yu Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chen Ling
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Kang Chen
- Department of Obstetrics and Gynecology and Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Dan Ye
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
8
|
Chène P. Direct Inhibition of the YAP : TEAD Interaction: An Unprecedented Drug Discovery Challenge. ChemMedChem 2024; 19:e202400361. [PMID: 38863297 DOI: 10.1002/cmdc.202400361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/13/2024]
Abstract
The Hippo pathway, which is key in organ morphogenesis, is frequently deregulated in cancer. The TEAD (TEA domain family member) transcription factors are the most distal elements of this pathway, and their activity is regulated by proteins such as YAP (Yes-associated protein). The identification of inhibitors of the YAP : TEAD interaction is one approach to develop novel anticancer drugs: the first clinical candidate (IAG933) preventing the association between these two proteins by direct competition has just been reported. The discovery of this molecule was particularly challenging because the interface between these two proteins is large (~3500 Å2 buried in complex formation) and made up of distinct contact areas. The most critical of these involves an omega-loop (Ω-loop), a secondary structure element rarely found in protein-protein interactions. This review summarizes how the knowledge gained from structure-function studies of the interaction between the Ω-loop of YAP and TEAD was used to devise the strategy to identify potent low-molecular weight compounds that show a pronounced anti-tumor effect.
Collapse
Affiliation(s)
- Patrick Chène
- Disease Area Oncology, Biomedical Research, CH-4056, Basel, Switzerland
- Novartis, WSJ 386 4.13.06, CH-4002, Basel, Switzerland
| |
Collapse
|
9
|
Pobbati AV, Burtscher A, Rajaram Siva N, Hallett A, Romigh T, Che K, Zhao B, Coker JA, Wang N, Stauffer SR, Rubin BP. CDK9 Inhibition by Dinaciclib Is a Therapeutic Vulnerability in Epithelioid Hemangioendothelioma. Clin Cancer Res 2024; 30:4179-4189. [PMID: 39052240 PMCID: PMC11398983 DOI: 10.1158/1078-0432.ccr-24-1097] [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: 04/08/2024] [Revised: 07/02/2024] [Accepted: 07/23/2024] [Indexed: 07/27/2024]
Abstract
PURPOSE There are no effective treatment options for patients with aggressive epithelioid hemangioendothelioma (EHE) driven by the TAZ-CAMTA1 (TC) fusion gene. Here, we aimed to understand the regulation of TC using pharmacologic tools and identify vulnerabilities that can potentially be exploited for the treatment of EHE. EXPERIMENTAL DESIGN TC is a transcriptional coregulator; we hypothesized that compounds that reduce TC nuclear levels, either through translocation of TC to the cytoplasm, or through degradation, would render TC less oncogenic. TC localization was monitored using immunofluorescence in an EHE tumor cell line. Two target-selective libraries were used to identify small molecules that reduce TC localization in the nucleus. The ability of the shortlisted hits to affect cell viability, apoptosis, and tumorigenesis was also evaluated. RESULTS Basal TC remained "immobile" in the nucleus; administration of cyclin-dependent kinase (CDK) inhibitors such as CGP60474 and dinaciclib (Dina) mobilized TC. "Mobile" TC shuttled between the nucleus and cytoplasm; however, it was eventually degraded through proteasomes. This dramatically suppressed the levels of TC-regulated transcripts and cell viability, promoted apoptosis, and reduced the area of metastatic lesions in the allograft model of EHE. We specifically identified that the inhibition of CDK9, a transcriptional CDK, destabilizes TC. CONCLUSIONS The CDK inhibitor Dina exhibited antitumorigenic properties both in vitro and in vivo in EHE models. Dina has been rigorously tested in clinical trials and displayed an acceptable toxicity profile. Therefore, there is a potential therapeutic window for repurposing Dina for the treatment of EHE.
Collapse
Affiliation(s)
- Ajaybabu V Pobbati
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Ashley Burtscher
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Nandini Rajaram Siva
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Andrea Hallett
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Todd Romigh
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Kepeng Che
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Bin Zhao
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Jesse A Coker
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Nancy Wang
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Shaun R Stauffer
- Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Brian P Rubin
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio
- Robert J. Tomsich Department of Pathology and Laboratory Medicine, Diagnostics Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| |
Collapse
|
10
|
Kulkarni A, Mohan V, Tang TT, Post L, Chan YC, Manning M, Thio N, Parker BL, Dawson MA, Rosenbluh J, Vissers JH, Harvey KF. Identification of resistance mechanisms to small-molecule inhibition of TEAD-regulated transcription. EMBO Rep 2024; 25:3944-3969. [PMID: 39103676 PMCID: PMC11387499 DOI: 10.1038/s44319-024-00217-3] [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: 06/27/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 08/07/2024] Open
Abstract
The Hippo tumor suppressor pathway controls transcription by regulating nuclear abundance of YAP and TAZ, which activate transcription with the TEAD1-TEAD4 DNA-binding proteins. Recently, several small-molecule inhibitors of YAP and TEADs have been reported, with some entering clinical trials for different cancers with Hippo pathway deregulation, most notably, mesothelioma. Using genome-wide CRISPR/Cas9 screens we reveal that mutations in genes from the Hippo, MAPK, and JAK-STAT signaling pathways all modulate the response of mesothelioma cell lines to TEAD palmitoylation inhibitors. By exploring gene expression programs of mutant cells, we find that MAPK pathway hyperactivation confers resistance to TEAD inhibition by reinstating expression of a subset of YAP/TAZ target genes. Consistent with this, combined inhibition of TEAD and the MAPK kinase MEK, synergistically blocks proliferation of multiple mesothelioma and lung cancer cell lines and more potently reduces the growth of patient-derived lung cancer xenografts in vivo. Collectively, we reveal mechanisms by which cells can overcome small-molecule inhibition of TEAD palmitoylation and potential strategies to enhance the anti-tumor activity of emerging Hippo pathway targeted therapies.
Collapse
Affiliation(s)
- Aishwarya Kulkarni
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Varshini Mohan
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Tracy T Tang
- Vivace Therapeutics Inc., San Mateo, CA, 94404, USA
| | - Leonard Post
- Vivace Therapeutics Inc., San Mateo, CA, 94404, USA
| | - Yih-Chih Chan
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Murray Manning
- Department of Biochemistry, and Biomedicine Discovery Institute, Monash University, Clayton, 3800, Australia
- Functional Genomics Platform, Monash University, Clayton, VIC, 3800, Australia
| | - Niko Thio
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Benjamin L Parker
- Department of Anatomy & Physiology, The University of Melbourne, Parkville, 3010, VIC, Australia
| | - Mark A Dawson
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Centre for Cancer Research and Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Joseph Rosenbluh
- Department of Biochemistry, and Biomedicine Discovery Institute, Monash University, Clayton, 3800, Australia
- Functional Genomics Platform, Monash University, Clayton, VIC, 3800, Australia
| | - Joseph Ha Vissers
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Centre for Cancer Research and Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Kieran F Harvey
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia.
- Department of Anatomy and Developmental Biology, and Biomedicine Discovery Institute, Monash University, Clayton, 3800, Australia.
| |
Collapse
|
11
|
Kim CL, Lim SB, Kim DH, Sim YE, Kang LJ, Park SJ, Kim H, Roh TH, Mo JS, Jeong HS. Regulation of Hippo-YAP signaling axis by Isoalantolactone suppresses tumor progression in cholangiocarcinoma. Transl Oncol 2024; 46:101971. [PMID: 38797019 PMCID: PMC11152753 DOI: 10.1016/j.tranon.2024.101971] [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: 01/18/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 05/29/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a devastating malignancy characterized by aggressive tumor growth and limited treatment options. Dysregulation of the Hippo signaling pathway and its downstream effector, Yes-associated protein (YAP), has been implicated in CCA development and progression. In this study, we investigated the effects of Isoalantolactone (IALT) on CCA cells to elucidate its effect on YAP activity and its potential clinical significance. Our findings demonstrate that IALT exerts cytotoxic effects, induces apoptosis, and modulates YAP signaling in SNU478 cells. We further confirmed the involvement of the canonical Hippo pathway by generating LATS1/LATS2 knockout cells, highlighting the dependence of IALT-mediated apoptosis and YAP phosphorylation on the Hippo-LATS signaling axis. In addition, IALT suppressed cell growth and migration, partially dependent on YAP-TEAD activity. These results provide insights into the therapeutic potential of targeting YAP in CCA and provide a rationale for developing of YAP-targeted therapies for this challenging malignancy.
Collapse
Affiliation(s)
- Cho-Long Kim
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon 16499, South Korea
| | - Su-Bin Lim
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon 16499, South Korea
| | - Dong Hyun Kim
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon 16499, South Korea
| | - Ye Eun Sim
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon 16499, South Korea
| | - Li-Jung Kang
- Three-Dimensional Immune System Imaging Core Facility, Ajou University, Suwon 16499, South Korea
| | - Su Jung Park
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan 50612, South Korea
| | - Hyungwoo Kim
- Division of Pharmacology, School of Korean Medicine, Pusan National University, Yangsan 50612, South Korea
| | - Tae Hoon Roh
- Department of Neurosurgery, Ajou University School of Medicine, Suwon 16499, South Korea
| | - Jung-Soon Mo
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon 16499, South Korea; Institute of Medical Science, Ajou University School of Medicine, Suwon 16499, South Korea.
| | - Han-Sol Jeong
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan 50612, South Korea.
| |
Collapse
|
12
|
Benton D, Yee Chow H, Karchugina S, Chernoff J. Synergistic effect of PAK and Hippo pathway inhibitor combination in NF2-deficient Schwannoma. PLoS One 2024; 19:e0305121. [PMID: 39083549 PMCID: PMC11290668 DOI: 10.1371/journal.pone.0305121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 05/23/2024] [Indexed: 08/02/2024] Open
Abstract
Neurofibromatosis type 2 is a genetic disorder that results in the formation and progressive growth of schwannomas, ependymomas, and/or meningiomas. The NF2 gene encodes the Merlin protein, which links cell cortical elements to the actin cytoskeleton and regulates a number of key enzymes including Group I p21-activated kinases (PAKs), the Hippo-pathway kinase LATS, and mTORC. While PAK1 and PAK2 directly bind Merlin and transmit proliferation and survival signals when Merlin is mutated or absent, inhibition of Group 1 PAKs alone has not proven sufficient to completely stop the growth of NF2-deficient meningiomas or schwannomas in vivo, suggesting the need for a second pathway inhibitor. As the Hippo pathway is also activated in NF2-deficient cells, several inhibitors of the Hippo pathway have recently been developed in the form of YAP-TEAD binding inhibitors. These inhibitors prevent activation of pro-proliferation and anti-apoptotic Hippo pathway effectors. In this study, we show that PAK inhibition slows cell proliferation while TEAD inhibition promotes apoptotic cell death. Finally, we demonstrate the efficacy of PAK and TEAD inhibitor combinations in several NF2-deficient Schwannoma cell lines.
Collapse
Affiliation(s)
- Dorothy Benton
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Hoi Yee Chow
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Sofiia Karchugina
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Jonathan Chernoff
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
13
|
Chapeau EA, Sansregret L, Galli GG, Chène P, Wartmann M, Mourikis TP, Jaaks P, Baltschukat S, Barbosa IAM, Bauer D, Brachmann SM, Delaunay C, Estadieu C, Faris JE, Furet P, Harlfinger S, Hueber A, Jiménez Núñez E, Kodack DP, Mandon E, Martin T, Mesrouze Y, Romanet V, Scheufler C, Sellner H, Stamm C, Sterker D, Tordella L, Hofmann F, Soldermann N, Schmelzle T. Direct and selective pharmacological disruption of the YAP-TEAD interface by IAG933 inhibits Hippo-dependent and RAS-MAPK-altered cancers. NATURE CANCER 2024; 5:1102-1120. [PMID: 38565920 PMCID: PMC11286534 DOI: 10.1038/s43018-024-00754-9] [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: 05/22/2023] [Accepted: 03/01/2024] [Indexed: 04/04/2024]
Abstract
The YAP-TEAD protein-protein interaction mediates YAP oncogenic functions downstream of the Hippo pathway. To date, available YAP-TEAD pharmacologic agents bind into the lipid pocket of TEAD, targeting the interaction indirectly via allosteric changes. However, the consequences of a direct pharmacological disruption of the interface between YAP and TEADs remain largely unexplored. Here, we present IAG933 and its analogs as potent first-in-class and selective disruptors of the YAP-TEAD protein-protein interaction with suitable properties to enter clinical trials. Pharmacologic abrogation of the interaction with all four TEAD paralogs resulted in YAP eviction from chromatin and reduced Hippo-mediated transcription and induction of cell death. In vivo, deep tumor regression was observed in Hippo-driven mesothelioma xenografts at tolerated doses in animal models as well as in Hippo-altered cancer models outside mesothelioma. Importantly this also extended to larger tumor indications, such as lung, pancreatic and colorectal cancer, in combination with RTK, KRAS-mutant selective and MAPK inhibitors, leading to more efficacious and durable responses. Clinical evaluation of IAG933 is underway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Daniel Bauer
- Novartis BioMedical Research, Basel, Switzerland
| | | | | | | | | | - Pascal Furet
- Novartis BioMedical Research, Basel, Switzerland
| | - Stefanie Harlfinger
- Novartis BioMedical Research, Basel, Switzerland
- AstraZeneca, Oncology R&D, Cambridge, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | - Francesco Hofmann
- Novartis BioMedical Research, Basel, Switzerland
- Pierre Fabre Group, R&D Medical Care, Toulouse, France
| | | | | |
Collapse
|
14
|
Cao Z, Hou Y, Zhao Z, Zhang H, Tian L, Zhang Y, Dong C, Guo F, Tan L, Han Y, Wang W, Jiao S, Tang Y, An L, Zhou Z. Reactivating Hippo by drug compounds to suppress gastric cancer and enhance chemotherapy sensitivity. J Biol Chem 2024; 300:107311. [PMID: 38657866 PMCID: PMC11126936 DOI: 10.1016/j.jbc.2024.107311] [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: 01/10/2024] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024] Open
Abstract
The Hippo signaling pathway plays an essential role in organ size control and tumorigenesis. Loss of Hippo signal and hyper-activation of the downstream oncogenic YAP signaling are commonly observed in various types of cancers. We previously identified STRN3-containing PP2A phosphatase as a negative regulator of MST1/2 kinases (i.e., Hippo) in gastric cancer (GC), opening the possibility of selectively targeting the PP2Aa-STRN3-MST1/2 axis to recover Hippo signaling against cancer. Here, we further discovered 1) disulfiram (DSF), an FDA-approved drug, which can similarly block the binding of STRN3 to PP2A core enzyme and 2) CX-6258 (CX), a chemical inhibitor, that can disrupt the interaction between STRN3 and MST1/2, both allowing reactivation of Hippo activity to inhibit GC. More importantly, we found these two compounds, via an MST1/2 kinase-dependent manner, inhibit DNA repair to sensitize GC towards chemotherapy. In addition, we identified thiram, a structural analog of DSF, can function similarly to inhibit cancer cell proliferation or enhance chemotherapy sensitivity. Interestingly, inclusion of copper ion enhanced such effects of DSF and thiram on GC treatment. Overall, this work demonstrated that pharmacological targeting of the PP2Aa-STRN3-MST1/2 axis by drug compounds can potently recover Hippo signal for tumor treatment.
Collapse
Affiliation(s)
- Zhifa Cao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Stomatology, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Tongji University Cancer Center, School of Medicine, Tongji University, Shanghai, China
| | - Yu Hou
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhangting Zhao
- Department of Stomatology, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Tongji University Cancer Center, School of Medicine, Tongji University, Shanghai, China
| | - Hui Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Luyang Tian
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yiming Zhang
- Department of Stomatology, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Tongji University Cancer Center, School of Medicine, Tongji University, Shanghai, China
| | - Chao Dong
- Department of Medical Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fenghua Guo
- Department of General Surgery, Hua'shan Hospital, Fudan University Shanghai Medical College, Shanghai, China
| | - Lijie Tan
- Department of Thoracic Surgery, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yi Han
- Department of Stomatology, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Tongji University Cancer Center, School of Medicine, Tongji University, Shanghai, China
| | - Wenjia Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shi Jiao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yang Tang
- Department of Stomatology, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Tongji University Cancer Center, School of Medicine, Tongji University, Shanghai, China.
| | - Liwei An
- Department of Stomatology, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Tongji University Cancer Center, School of Medicine, Tongji University, Shanghai, China.
| | - Zhaocai Zhou
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China; Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
15
|
Guo S, Hu X, Cotton JL, Ma L, Li Q, Cui J, Wang Y, Thakare RP, Tao Z, Ip YT, Wu X, Wang J, Mao J. VGLL2 and TEAD1 fusion proteins drive YAP/TAZ-independent transcription and tumorigenesis by engaging p300. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.01.592016. [PMID: 38746415 PMCID: PMC11092657 DOI: 10.1101/2024.05.01.592016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Studies on Hippo pathway regulation of tumorigenesis largely center on YAP and TAZ, the transcriptional co-regulators of TEAD. Here, we present an oncogenic mechanism involving VGLL and TEAD fusions that is Hippo pathway-related but YAP/TAZ-independent. We characterize two recurrent fusions, VGLL2-NCOA2 and TEAD1-NCOA2, recently identified in spindle cell rhabdomyosarcoma. We demonstrate that, in contrast to VGLL2 and TEAD1, the fusion proteins are strong activators of TEAD-dependent transcription, and their function does not require YAP/TAZ. Furthermore, we identify that VGLL2 and TEAD1 fusions engage specific epigenetic regulation by recruiting histone acetyltransferase p300 to control TEAD-mediated transcriptional and epigenetic landscapes. We showed that small molecule p300 inhibition can suppress fusion proteins-induced oncogenic transformation both in vitro and in vivo. Overall, our study reveals a molecular basis for VGLL involvement in cancer and provides a framework for targeting tumors carrying VGLL, TEAD, or NCOA translocations.
Collapse
Affiliation(s)
- Susu Guo
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No 241, West Huaihai Road, Shanghai, P. R., 200030, China
| | - Xiaodi Hu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, 01605, USA
| | - Jennifer L. Cotton
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, 01605, USA
| | - Lifang Ma
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No 241, West Huaihai Road, Shanghai, P. R., 200030, China
| | - Qi Li
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, 01605, USA
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, 01605, USA
| | - Jiangtao Cui
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No 241, West Huaihai Road, Shanghai, P. R., 200030, China
| | - Yongjie Wang
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No 241, West Huaihai Road, Shanghai, P. R., 200030, China
| | - Ritesh P. Thakare
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, 01605, USA
| | - Zhipeng Tao
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, 01605, USA
| | - Y. Tony Ip
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, 01605, USA
| | - Xu Wu
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, 01605, USA
| | - Jiayi Wang
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No 241, West Huaihai Road, Shanghai, P. R., 200030, China
| | - Junhao Mao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, 01605, USA
| |
Collapse
|
16
|
Kim HJ, Choi Y, Lee Y, Hwangbo M, Kim J. OTUD6A orchestrates complex modulation of TEAD4-mediated transcriptional programs. FEBS Lett 2024; 598:1045-1060. [PMID: 38594215 DOI: 10.1002/1873-3468.14861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/28/2024] [Accepted: 03/02/2024] [Indexed: 04/11/2024]
Abstract
TEAD transcription factors play a central role in the Hippo signaling pathway. In this study, we focused on transcriptional enhancer factor TEF-3 (TEAD4), exploring its regulation by the deubiquitinase OTU domain-containing protein 6A (OTUD6A). We identified OTUD6A as a TEAD4-interacting deubiquitinase, positively influencing TEAD-driven transcription without altering TEAD4 stability. Structural analyses revealed specific interaction domains: the N-terminal domain of OTUD6A and the YAP-binding domain of TEAD4. Functional assays demonstrated the positive impact of OTUD6A on the transcription of YAP-TEAD target genes. Despite no impact on TEAD4 nuclear localization, OTUD6A selectively modulated nuclear interactions, enhancing YAP-TEAD4 complex formation while suppressing VGLL4 (transcription cofactor vestigial-like protein 4)-TEAD4 interaction. Critically, OTUD6A facilitated YAP-TEAD4 complex binding to target gene promoters. Our study unveils the regulatory landscape of OTUD6A on TEAD4, providing insights into diseases regulated by YAP-TEAD complexes.
Collapse
Affiliation(s)
- Hyo Jin Kim
- Department of Life Sciences, Sogang University, Seoul, Korea
| | - Yunsik Choi
- Department of Life Sciences, Sogang University, Seoul, Korea
| | - Yuri Lee
- Department of Life Sciences, Sogang University, Seoul, Korea
| | - Mi Hwangbo
- Department of Life Sciences, Sogang University, Seoul, Korea
| | - Jongchan Kim
- Department of Life Sciences, Sogang University, Seoul, Korea
| |
Collapse
|
17
|
Sommerauer C, Gallardo-Dodd CJ, Savva C, Hases L, Birgersson M, Indukuri R, Shen JX, Carravilla P, Geng K, Nørskov Søndergaard J, Ferrer-Aumatell C, Mercier G, Sezgin E, Korach-André M, Petersson C, Hagström H, Lauschke VM, Archer A, Williams C, Kutter C. Estrogen receptor activation remodels TEAD1 gene expression to alleviate hepatic steatosis. Mol Syst Biol 2024; 20:374-402. [PMID: 38459198 PMCID: PMC10987545 DOI: 10.1038/s44320-024-00024-x] [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: 09/14/2023] [Revised: 02/10/2024] [Accepted: 02/13/2024] [Indexed: 03/10/2024] Open
Abstract
Sex-based differences in obesity-related hepatic malignancies suggest the protective roles of estrogen. Using a preclinical model, we dissected estrogen receptor (ER) isoform-driven molecular responses in high-fat diet (HFD)-induced liver diseases of male and female mice treated with or without an estrogen agonist by integrating liver multi-omics data. We found that selective ER activation recovers HFD-induced molecular and physiological liver phenotypes. HFD and systemic ER activation altered core liver pathways, beyond lipid metabolism, that are consistent between mice and primates. By including patient cohort data, we uncovered that ER-regulated enhancers govern central regulatory and metabolic genes with clinical significance in metabolic dysfunction-associated steatotic liver disease (MASLD) patients, including the transcription factor TEAD1. TEAD1 expression increased in MASLD patients, and its downregulation by short interfering RNA reduced intracellular lipid content. Subsequent TEAD small molecule inhibition improved steatosis in primary human hepatocyte spheroids by suppressing lipogenic pathways. Thus, TEAD1 emerged as a new therapeutic candidate whose inhibition ameliorates hepatic steatosis.
Collapse
Affiliation(s)
- Christian Sommerauer
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Carlos J Gallardo-Dodd
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Christina Savva
- Department of Medicine, Integrated Cardio Metabolic Center, Karolinska Institute, Huddinge, Sweden
| | - Linnea Hases
- Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Madeleine Birgersson
- Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Rajitha Indukuri
- Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Joanne X Shen
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Pablo Carravilla
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
- Department of Women's and Children's Health, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Keyi Geng
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Jonas Nørskov Søndergaard
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Clàudia Ferrer-Aumatell
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Grégoire Mercier
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Erdinc Sezgin
- Department of Women's and Children's Health, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Marion Korach-André
- Department of Medicine, Integrated Cardio Metabolic Center, Karolinska Institute, Huddinge, Sweden
| | - Carl Petersson
- Department of Drug Metabolism and Pharmacokinetics, The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Hannes Hagström
- Department of Medicine Huddinge, Karolinska Institute, Huddinge, Sweden
- Division of Hepatology, Department of Upper GI Diseases, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - Amena Archer
- Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Cecilia Williams
- Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Claudia Kutter
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden.
| |
Collapse
|
18
|
Kumar R, Hong W. Hippo Signaling at the Hallmarks of Cancer and Drug Resistance. Cells 2024; 13:564. [PMID: 38607003 PMCID: PMC11011035 DOI: 10.3390/cells13070564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/11/2024] [Accepted: 03/20/2024] [Indexed: 04/13/2024] Open
Abstract
Originally identified in Drosophila melanogaster in 1995, the Hippo signaling pathway plays a pivotal role in organ size control and tumor suppression by inhibiting proliferation and promoting apoptosis. Large tumor suppressors 1 and 2 (LATS1/2) directly phosphorylate the Yki orthologs YAP (yes-associated protein) and its paralog TAZ (also known as WW domain-containing transcription regulator 1 [WWTR1]), thereby inhibiting their nuclear localization and pairing with transcriptional coactivators TEAD1-4. Earnest efforts from many research laboratories have established the role of mis-regulated Hippo signaling in tumorigenesis, epithelial mesenchymal transition (EMT), oncogenic stemness, and, more recently, development of drug resistances. Hippo signaling components at the heart of oncogenic adaptations fuel the development of drug resistance in many cancers for targeted therapies including KRAS and EGFR mutants. The first U.S. food and drug administration (US FDA) approval of the imatinib tyrosine kinase inhibitor in 2001 paved the way for nearly 100 small-molecule anti-cancer drugs approved by the US FDA and the national medical products administration (NMPA). However, the low response rate and development of drug resistance have posed a major hurdle to improving the progression-free survival (PFS) and overall survival (OS) of cancer patients. Accumulating evidence has enabled scientists and clinicians to strategize the therapeutic approaches of targeting cancer cells and to navigate the development of drug resistance through the continuous monitoring of tumor evolution and oncogenic adaptations. In this review, we highlight the emerging aspects of Hippo signaling in cross-talk with other oncogenic drivers and how this information can be translated into combination therapy to target a broad range of aggressive tumors and the development of drug resistance.
Collapse
Affiliation(s)
- Ramesh Kumar
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology, and Research), Singapore 138673, Singapore;
| | | |
Collapse
|
19
|
Imura T, Mitsuhara T, Horie N. Characteristics of MicroRNA Expression Depending on the Presence or Absence of Meningioma in Patients with Neurofibromatosis Type 2: A Secondary Analysis. Neurol Med Chir (Tokyo) 2024; 64:116-122. [PMID: 38267057 PMCID: PMC10992986 DOI: 10.2176/jns-nmc.2023-0200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/24/2023] [Indexed: 01/26/2024] Open
Abstract
Meningioma is the second most frequent tumor in patients with neurofibromatosis type 2 (NF2). The presence of meningioma is believed to be a negative prognostic marker in these patients. However, the molecular mechanisms involved in the tumorigenesis of NF2-associated meningioma are not well characterized. Epigenetic regulation, including microRNAs (miRNAs), may be involved in the development of different tumor types in patients with NF2. The objective of this study is to explore the different characteristics of serum miRNA expression depending on the presence or absence of meningioma in patients with NF2. Nine patients with NF2 who were treated at the Department of Neurosurgery, Hiroshima University Hospital, were included. Total RNA (including small RNAs) was extracted from serum samples for the preparation of a small RNA library for next-generation sequencing analysis. Differentially expressed miRNAs (DEMs) were analyzed using the DESeq2 package to compare the characteristic miRNA expression profiles of patients with and without meningioma. In small RNA sequencing analysis, out of a total of 1,879 miRNAs registered in the database, the expressions of 657 miRNAs were observed. In DEM analysis, the expressions of four miRNAs, namely, hsa-miR-664b, hsa-miR-7706, hsa-miR-590, and hsa-miR-6513, were downregulated in patients with NF2 with meningioma compared with patients with NF2 without meningioma. Hsa-miR-193a was identified as the only upregulated miRNA in patients with NF2 with meningioma. In conclusion, we identified different circulating miRNA expression characteristics depending on the presence or absence of meningioma in patients with NF2.
Collapse
Affiliation(s)
- Takeshi Imura
- Department of Rehabilitation, Faculty of Health Sciences, Hiroshima Cosmopolitan University
| | - Takafumi Mitsuhara
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Nobutaka Horie
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University
| |
Collapse
|
20
|
Zou H, Luo J, Guo Y, Deng L, Zeng L, Pan Y, Li P. Tyrosine phosphorylation-mediated YAP1-TFAP2A interactions coordinate transcription and trastuzumab resistance in HER2+ breast cancer. Drug Resist Updat 2024; 73:101051. [PMID: 38219531 DOI: 10.1016/j.drup.2024.101051] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 12/31/2023] [Accepted: 01/05/2024] [Indexed: 01/16/2024]
Abstract
Trastuzumab resistance in HER2+ breast cancer (BC) is the major reason leading to poor prognosis of BC patients. Oncogenic gene overexpression or aberrant activation of tyrosine kinase SRC is identified to be the key modulator of trastuzumab response. However, the detailed regulatory mechanisms underlying SRC activation-associated trastuzumab resistance remain poorly understood. In the present study, we discover that SRC-mediated YAP1 tyrosine phosphorylation facilitates its interaction with transcription factor AP-2 alpha (activating enhancer binding protein 2 alpha, TFAP2A), which in turn promotes YAP1/TEAD-TFAP2A (YTT) complex-associated transcriptional outputs, thereby conferring trastuzumab resistance in HER2+ BC. Inhibition of SRC kinase activity or disruption of YTT complex sensitizes cells to trastuzumab treatment in vitro and in vivo. Additionally, we also identify YTT complex co-occupies the regulatory regions of a series of genes related to trastuzumab resistance and directly regulates their transcriptions, including EGFR, HER2, H19 and CTGF. Moreover, YTT-mediated transcriptional regulation is coordinated by SRC kinase activity. Taken together, our study reveals that SRC-mediated YTT complex formation and transcriptions are responsible for multiple mechanisms associated with trastuzumab resistance. Therefore, targeting HER2 signaling in combination with the inhibition of YTT-associated transcriptional outputs could serve as the treatment strategy to overcome trastuzumab resistance caused by SRC activation.
Collapse
Affiliation(s)
- Hailin Zou
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China
| | - Juan Luo
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China
| | - Yibo Guo
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China
| | - Liang Deng
- Department of General Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China
| | - Leli Zeng
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China
| | - Yihang Pan
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China.
| | - Peng Li
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China.
| |
Collapse
|
21
|
Tape CJ. Plastic persisters: revival stem cells in colorectal cancer. Trends Cancer 2024; 10:185-195. [PMID: 38071119 DOI: 10.1016/j.trecan.2023.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 03/16/2024]
Abstract
Colorectal cancer (CRC) is traditionally considered to be a genetically driven disease. However, nongenetic plasticity has recently emerged as a major driver of tumour initiation, metastasis, and therapy response in CRC. Central to these processes is a recently discovered cell type, the revival colonic stem cell (revCSC). In contrast to traditional proliferative CSCs (proCSCs), revCSCs prioritise survival over propagation. revCSCs play an essential role in primary tumour formation, metastatic dissemination, and nongenetic chemoresistance. Current evidence suggests that CRC tumours leverage intestinal stem cell plasticity to both proliferate (via proCSCs) when unchallenged and survive (via revCSCs) in response to cell-extrinsic pressures. Although revCSCs likely represent a major source of therapeutic failure in CRC, our increasing knowledge of this important stem cell fate provides novel opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Christopher J Tape
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London, WC1E 6DD, UK.
| |
Collapse
|
22
|
Wang Y, Yu H, Cen Z, Zhu Y, Wu W. Drug targets regulate systemic metabolism and provide new horizons to treat nonalcoholic steatohepatitis. Metabol Open 2024; 21:100267. [PMID: 38187470 PMCID: PMC10770762 DOI: 10.1016/j.metop.2023.100267] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 12/06/2023] [Accepted: 12/12/2023] [Indexed: 01/09/2024] Open
Abstract
Nonalcoholic steatohepatitis (NASH), is the advanced stage of nonalcoholic fatty liver disease (NAFLD) with rapidly rising global prevalence. It is featured with severe hepatocyte apoptosis, inflammation and hepatic lipogenesis. The drugs directly targeting the processes of steatosis, inflammation and fibrosis are currently under clinical investigation. Nevertheless, the long-term ineffectiveness and remarkable adverse effects are well documented, and new concepts are required to tackle with the root causes of NASH progression. We critically assess the recently validated drug targets that regulate the systemic metabolism to ameliorate NASH. Thermogenesis promoted by mitochondrial uncouplers restores systemic energy expenditure. Furthermore, regulation of mitochondrial proteases and proteins that are pivotal for intracellular metabolic homeostasis normalize mitochondrial function. Secreted proteins also improve systemic metabolism, and NASH is ameliorated by agonizing receptors of secreted proteins with small molecules. We analyze the drug design, the advantages and shortcomings of these novel drug candidates. Meanwhile, the structural modification of current NASH therapeutics significantly increased their selectivity, efficacy and safety. Furthermore, the arising CRISPR-Cas9 screen strategy on liver organoids has enabled the identification of new genes that mediate lipid metabolism, which may serve as promising drug targets. In summary, this article discusses the in-depth novel mechanisms and the multidisciplinary approaches, and they provide new horizons to treat NASH.
Collapse
Affiliation(s)
- Yibing Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai, 200438, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, China
| | - Hanhan Yu
- School of Kinesiology, Shanghai University of Sport, Shanghai, 200438, China
| | - Zhipeng Cen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China
| | - Yutong Zhu
- School of Kinesiology, Shanghai University of Sport, Shanghai, 200438, China
| | - Wenyi Wu
- School of Kinesiology, Shanghai University of Sport, Shanghai, 200438, China
| |
Collapse
|
23
|
Kanai R, Norton E, Stern P, Hynes RO, Lamar JM. Identification of a Gene Signature That Predicts Dependence upon YAP/TAZ-TEAD. Cancers (Basel) 2024; 16:852. [PMID: 38473214 PMCID: PMC10930532 DOI: 10.3390/cancers16050852] [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: 12/27/2023] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 03/14/2024] Open
Abstract
Targeted therapies are effective cancer treatments when accompanied by accurate diagnostic tests that can help identify patients that will respond to those therapies. The YAP/TAZ-TEAD axis is activated and plays a causal role in several cancer types, and TEAD inhibitors are currently in early-phase clinical trials in cancer patients. However, a lack of a reliable way to identify tumors with YAP/TAZ-TEAD activation for most cancer types makes it difficult to determine which tumors will be susceptible to TEAD inhibitors. Here, we used a combination of RNA-seq and bioinformatic analysis of metastatic melanoma cells to develop a YAP/TAZ gene signature. We found that the genes in this signature are TEAD-dependent in several melanoma cell lines, and that their expression strongly correlates with YAP/TAZ activation in human melanomas. Using DepMap dependency data, we found that this YAP/TAZ signature was predictive of melanoma cell dependence upon YAP/TAZ or TEADs. Importantly, this was not limited to melanoma because this signature was also predictive when tested on a panel of over 1000 cancer cell lines representing numerous distinct cancer types. Our results suggest that YAP/TAZ gene signatures like ours may be effective tools to predict tumor cell dependence upon YAP/TAZ-TEAD, and thus potentially provide a means to identify patients likely to benefit from TEAD inhibitors.
Collapse
Affiliation(s)
- Ryan Kanai
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (R.K.); (E.N.)
| | - Emily Norton
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (R.K.); (E.N.)
| | - Patrick Stern
- Koch Institute for Integrative Cancer Research, at Massachusetts Institute of Technology, Cambridge, MA 02139, USA;
| | - Richard O. Hynes
- Department of Biology, Koch Institute for Integrative Cancer Research, and Howard Hughes Medical Institute, at Massachusetts Institute of Technology, Cambridge, MA 02139, USA;
| | - John M. Lamar
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (R.K.); (E.N.)
| |
Collapse
|
24
|
Zhou Y, Wang D, Zhou L, Zhou N, Wang Z, Chen J, Pang R, Fu H, Huang Q, Dong F, Cheng H, Zhang H, Tang K, Ma J, Lv J, Cheng T, Fiskesund R, Zhang X, Huang B. Cell softness renders cytotoxic T lymphocytes and T leukemic cells resistant to perforin-mediated killing. Nat Commun 2024; 15:1405. [PMID: 38360940 PMCID: PMC10869718 DOI: 10.1038/s41467-024-45750-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: 03/12/2023] [Accepted: 02/03/2024] [Indexed: 02/17/2024] Open
Abstract
Mechanical force contributes to perforin pore formation at immune synapses, thus facilitating the cytotoxic T lymphocytes (CTL)-mediated killing of tumor cells in a unidirectional fashion. How such mechanical cues affect CTL evasion of perforin-mediated autolysis remains unclear. Here we show that activated CTLs use their softness to evade perforin-mediated autolysis, which, however, is shared by T leukemic cells to evade CTL killing. Downregulation of filamin A is identified to induce softness via ZAP70-mediated YAP Y357 phosphorylation and activation. Despite the requirements of YAP in both cell types for softness induction, CTLs are more resistant to YAP inhibitors than malignant T cells, potentially due to the higher expression of the drug-resistant transporter, MDR1, in CTLs. As a result, moderate inhibition of YAP stiffens malignant T cells but spares CTLs, thus allowing CTLs to cytolyze malignant cells without autolysis. Our findings thus hint a mechanical force-based immunotherapeutic strategy against T cell leukemia.
Collapse
Affiliation(s)
- Yabo Zhou
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China
| | - Dianheng Wang
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China
| | - Li Zhou
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China
| | - Nannan Zhou
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China
| | - Zhenfeng Wang
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China
| | - Jie Chen
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China
| | - Ruiyang Pang
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China
| | - Haixia Fu
- Peking University People's Hospital, National Clinical Research Center for Hematologic Disease; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, Beijing, China
| | - Qiusha Huang
- Peking University People's Hospital, National Clinical Research Center for Hematologic Disease; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, Beijing, China
| | - Fang Dong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Hui Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Huafeng Zhang
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Tang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingwei Ma
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiadi Lv
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Roland Fiskesund
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Xiaohui Zhang
- Peking University People's Hospital, National Clinical Research Center for Hematologic Disease; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, Beijing, China.
| | - Bo Huang
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China.
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
25
|
Deng Y, Patel N, Ding S, Zhang H. Mechanism study of serum extracellular nano-vesicles miR-412-3p targeting regulation of TEAD1 in promoting malignant biological behavior of sub-centimeter lung nodules. Cancer Biomark 2024; 41:69-82. [PMID: 39269825 PMCID: PMC11495320 DOI: 10.3233/cbm-240137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/31/2024] [Indexed: 09/15/2024]
Abstract
OBJECTIVE To investigate the impact and potential mechanisms of serum extracellular nano-vesicles (sEVs) miR-412-3p released from sub-centimeter lung nodules with a diameter of ⩽ 10 mm on the malignant biological function of micro-nodular lung cancer (mnLC). METHODS A total of 87 participants were included and divided into a mnLC group (n= 30), a benign lung nodule (BLN) group (n= 27), and a healthy people control group (n= 30). Transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA) and Western blot (WB) were used to measure the morphological characteristics and surface markers of sEVs. In vitro analysis, real-time quantitative polymerase chain reaction (RT-qPCR), CCK-8 cell proliferation assay, clone formation assay, Transwell, stem cell sphere-forming assay, and WB assay were conducted to verify the effect of miR-412-3p/TEAD1 signaling axis on the biological function of lung cancer cells through, respectively. Further validation was conducted using the serum sEVs of the participants. RESULTS The expression level of sEVs-miR-412-3p in the mnLC group was significantly higher than that in the BLN and healthy groups (P< 0.01). In lung cancer cell lines, miR-412-3p can negatively regulate the targeted gene TEAD1. The miR-412-3p/TEAD1 signaling axis is involved in promoting the EMT signaling pathway and regulating the malignant biological functions of lung cancer cell proliferation, migration, and stemness (P< 0.05). In addition, sEVs in the mnLC group significantly promoted lung cancer cell proliferation, migration, and stemness compared to the BLN and healthy groups, inhibited the expression of E-cadherin and TEAD1 in lung cancer cells, and promoted the expression of N-cadherin and Vimentin (P< 0.05). CONCLUSION sEVs-miR-412-3p could promote the biological process of EMT, and lead to the occurrence of malignant biological behavior in sub-centimeter lung nodules. This provides evidence for the miR-412-3p/TEAD1 signaling axis as a potential therapeutic target for mnLC.
Collapse
Affiliation(s)
- Yuxia Deng
- Department of Oncology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Nishant Patel
- Department of Oncology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
- Department of Cardiothoracic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Shuang Ding
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Haijun Zhang
- Department of Oncology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| |
Collapse
|
26
|
Emon B, Joy MSH, Lalonde L, Ghrayeb A, Doha U, Ladehoff L, Brockstein R, Saengow C, Ewoldt RH, Saif MTA. Nuclear deformation regulates YAP dynamics in cancer associated fibroblasts. Acta Biomater 2024; 173:93-108. [PMID: 37977292 PMCID: PMC10848212 DOI: 10.1016/j.actbio.2023.11.015] [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: 07/20/2023] [Revised: 11/04/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023]
Abstract
Cells cultured on stiff 2D substrates exert high intracellular force, resulting in mechanical deformation of their nuclei. This nuclear deformation (ND) plays a crucial role in the transport of Yes Associated Protein (YAP) from the cytoplasm to the nucleus. However, cells in vivo are in soft 3D environment with potentially much lower intracellular forces. Whether and how cells may deform their nuclei in 3D for YAP localization remains unclear. Here, by culturing human colon cancer associated fibroblasts (CAFs) on 2D, 2.5D, and 3D substrates, we differentiated the effects of stiffness, force, and ND on YAP localization. We found that nuclear translocation of YAP depends on the degree of ND irrespective of dimensionality, stiffness and total force. ND induced by the perinuclear force, not the total force, and nuclear membrane curvature correlate strongly with YAP activation. Immunostained slices of human tumors further supported the association between ND and YAP nuclear localization, suggesting ND as a potential biomarker for YAP activation in tumors. Additionally, we conducted quantitative analysis of the force dynamics of CAFs on 2D substrates to construct a stochastic model of YAP kinetics. This model revealed that the probability of YAP nuclear translocation, as well as the residence time in the nucleus follow a power law. This study provides valuable insights into the regulatory mechanisms governing YAP dynamics and highlights the significance of threshold activation in YAP localization. STATEMENT OF SIGNIFICANCE: Yes Associated Protein (YAP), a transcription cofactor, has been identified as one of the drivers of cancer progression. High tumor stiffness is attributed to driving YAP to the nucleus, wherein it activates pro-metastatic genes. Here we show, using cancer associated fibroblasts, that YAP translocation to the nucleus depends on the degree of nuclear deformation, irrespective of stiffness. We also identified that perinuclear force induced membrane curvature correlates strongly with YAP nuclear transport. A novel stochastic model of YAP kinetics unveiled a power law relationship between the activation threshold and persistence time of YAP in the nucleus. Overall, this study provides novel insights into the regulatory mechanisms governing YAP dynamics and the probability of activation that is of immense clinical significance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Chaimongkol Saengow
- Mechanical Science & Engineering; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign
| | - Randy H Ewoldt
- Mechanical Science & Engineering; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign
| | - M Taher A Saif
- Mechanical Science & Engineering; Bioengineering; Cancer Center at Illinois.
| |
Collapse
|
27
|
Lei L, Yang J, Peng H, Huang R, Liang L, Liu R, Li J. The Prognostic Significance of the TEAD4 in Hepatocellular Carcinoma. Int J Gen Med 2023; 16:6005-6013. [PMID: 38144438 PMCID: PMC10748864 DOI: 10.2147/ijgm.s440973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/04/2023] [Indexed: 12/26/2023] Open
Abstract
Background Abnormal expression of genes causes tumorigenesis, tumor progression, and poor prognosis in hepatocellular carcinoma (HCC). Therefore, the aims of this study were to explore the transcription enhancer domain factor 4 (TEAD4) in patients with liver cancer and its relationship with prognosis. Methods HTSeq-FPKM data and corresponding clinical data of HCC patients were obtained from The Cancer Genome Atlas (TCGA). Difference in TEAD4 expression between normal and tumor and the correlation with clinical characteristics were analyzed by the chi-squared test based on UALCAN. HepG2 cell lines were used to study the effect of TEAD4 on HCC cell lines. The expression and clinical significance of TEAD4 in HCC were detected in clinical cases. Results The transcription and post-transcription levels of TEAD4 were higher in HCC tumors than normal illustrated different expressed transcription of TEAD4 in gender, nodal metastasis status, tumor grades, and individual cancer stages. The high TEAD4 expression was significantly associated with tumor grades. The high expression of TEAD4 was significantly correlated to shorter 2-5 years overall survival. Inhibition of TEAD4 expression in HepG2 cells resulted in significantly decreased cell proliferation and invasion. Conclusion TEAD4 was identified as an independent prognostic factor, and inhibition of TEAD4 expression in HepG2 cells resulted in significantly decreased cell proliferation and invasion.
Collapse
Affiliation(s)
- Liping Lei
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, People’s Republic of China
- Department of Geriatric Medicine, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, People’s Republic of China
| | - Jingjing Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, People’s Republic of China
| | - Hao Peng
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, People’s Republic of China
| | - Ruiyan Huang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, People’s Republic of China
| | - Lichun Liang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, People’s Republic of China
| | - Ruifang Liu
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, People’s Republic of China
| | - Jiangfa Li
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, People’s Republic of China
| |
Collapse
|
28
|
Benton D, Chernoff J. RHOA drivers take alternate routes in gastric cancer. Sci Signal 2023; 16:eadk9171. [PMID: 38113334 DOI: 10.1126/scisignal.adk9171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 11/17/2023] [Indexed: 12/21/2023]
Abstract
Oncogenic small guanosine triphosphatases (GTPases) are often characterized by a limited set of activating mutations that affect their intrinsic biochemical function, but RHOA-which is frequently mutated in gastric cancer-appears not to have read the instruction manual. Having previously characterized the Y42C RHOA mutation in gastric cancer, in this issue of Science Signaling, Schaefer et al. take on the slightly less common L57V mutation and find that individual RHOA mutations can have different and unpredictable signaling outcomes.
Collapse
Affiliation(s)
- Dorothy Benton
- Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA 19111, USA
| | - Jonathan Chernoff
- Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA 19111, USA
| |
Collapse
|
29
|
Mukhopadhyay S, Huang HY, Lin Z, Ranieri M, Li S, Sahu S, Liu Y, Ban Y, Guidry K, Hu H, Lopez A, Sherman F, Tan YJ, Lee YT, Armstrong AP, Dolgalev I, Sahu P, Zhang T, Lu W, Gray NS, Christensen JG, Tang TT, Velcheti V, Khodadadi-Jamayran A, Wong KK, Neel BG. Genome-Wide CRISPR Screens Identify Multiple Synthetic Lethal Targets That Enhance KRASG12C Inhibitor Efficacy. Cancer Res 2023; 83:4095-4111. [PMID: 37729426 PMCID: PMC10841254 DOI: 10.1158/0008-5472.can-23-2729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 09/22/2023]
Abstract
Non-small lung cancers (NSCLC) frequently (∼30%) harbor KRAS driver mutations, half of which are KRASG12C. KRAS-mutant NSCLC with comutated STK11 and/or KEAP1 is particularly refractory to conventional, targeted, and immune therapy. Development of KRASG12C inhibitors (G12Ci) provided a major therapeutic advance, but resistance still limits their efficacy. To identify genes whose deletion augments efficacy of the G12Cis adagrasib (MRTX-849) or adagrasib plus TNO155 (SHP2i), we performed genome-wide CRISPR/Cas9 screens on KRAS/STK11-mutant NSCLC lines. Recurrent, potentially targetable, synthetic lethal (SL) genes were identified, including serine-threonine kinases, tRNA-modifying and proteoglycan synthesis enzymes, and YAP/TAZ/TEAD pathway components. Several SL genes were confirmed by siRNA/shRNA experiments, and the YAP/TAZ/TEAD pathway was extensively validated in vitro and in mice. Mechanistic studies showed that G12Ci treatment induced gene expression of RHO paralogs and activators, increased RHOA activation, and evoked ROCK-dependent nuclear translocation of YAP. Mice and patients with acquired G12Ci- or G12Ci/SHP2i-resistant tumors showed strong overlap with SL pathways, arguing for the relevance of the screen results. These findings provide a landscape of potential targets for future combination strategies, some of which can be tested rapidly in the clinic. SIGNIFICANCE Identification of synthetic lethal genes with KRASG12C using genome-wide CRISPR/Cas9 screening and credentialing of the ability of TEAD inhibition to enhance KRASG12C efficacy provides a roadmap for combination strategies. See related commentary by Johnson and Haigis, p. 4005.
Collapse
Affiliation(s)
- Suman Mukhopadhyay
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Hsin-Yi Huang
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Ziyan Lin
- Applied Bioinformatics Laboratories, Office of Science and Research, New York University Grossman School of Medicine, New York, United States
| | - Michela Ranieri
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Shuai Li
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Soumyadip Sahu
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Yingzhuo Liu
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Yi Ban
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Kayla Guidry
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Hai Hu
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Alfonso Lopez
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Fiona Sherman
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Yi Jer Tan
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Yeuan Ting Lee
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Amanda P. Armstrong
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Igor Dolgalev
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Priyanka Sahu
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Tinghu Zhang
- Department of Chemical and Systems Biology, ChEM-H, Stanford Cancer Institute, School of Medicine, Stanford University, California, United States
| | - Wenchao Lu
- Department of Chemical and Systems Biology, ChEM-H, Stanford Cancer Institute, School of Medicine, Stanford University, California, United States
| | - Nathanael S. Gray
- Department of Chemical and Systems Biology, ChEM-H, Stanford Cancer Institute, School of Medicine, Stanford University, California, United States
| | | | - Tracy T. Tang
- Vivace Therapeutics, Inc., San Mateo, California, United States
| | - Vamsidhar Velcheti
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Alireza Khodadadi-Jamayran
- Applied Bioinformatics Laboratories, Office of Science and Research, New York University Grossman School of Medicine, New York, United States
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Benjamin G. Neel
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| |
Collapse
|
30
|
Edwards AC, Stalnecker CA, Morales AJ, Taylor KE, Klomp JE, Klomp JA, Waters AM, Sudhakar N, Hallin J, Tang TT, Olson P, Post L, Christensen JG, Cox AD, Der CJ. TEAD Inhibition Overcomes YAP1/TAZ-Driven Primary and Acquired Resistance to KRASG12C Inhibitors. Cancer Res 2023; 83:4112-4129. [PMID: 37934103 PMCID: PMC10821578 DOI: 10.1158/0008-5472.can-23-2994] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/01/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
Primary/intrinsic and treatment-induced acquired resistance limit the initial response rate to and long-term efficacy of direct inhibitors of the KRASG12C mutant in cancer. To identify potential mechanisms of resistance, we applied a CRISPR/Cas9 loss-of-function screen and observed loss of multiple components of the Hippo tumor suppressor pathway, which acts to suppress YAP1/TAZ-regulated gene transcription. YAP1/TAZ activation impaired the antiproliferative and proapoptotic effects of KRASG12C inhibitor (G12Ci) treatment in KRASG12C-mutant cancer cell lines. Conversely, genetic suppression of YAP1/WWTR1 (TAZ) enhanced G12Ci sensitivity. YAP1/TAZ activity overcame KRAS dependency through two distinct TEAD transcription factor-dependent mechanisms, which phenocopy KRAS effector signaling. First, TEAD stimulated ERK-independent transcription of genes normally regulated by ERK (BIRC5, CDC20, ECT2, FOSL1, and MYC) to promote progression through the cell cycle. Second, TEAD caused activation of PI3K-AKT-mTOR signaling to overcome apoptosis. G12Ci treatment-induced acquired resistance was also caused by YAP1/TAZ-TEAD activation. Accordingly, concurrent treatment with pharmacologic inhibitors of TEAD synergistically enhanced KRASG12C inhibitor antitumor activity in vitro and prolonged tumor suppression in vivo. In summary, these observations reveal YAP1/TAZ-TEAD signaling as a crucial driver of primary and acquired resistance to KRAS inhibition and support the use of TEAD inhibitors to enhance the antitumor efficacy of KRAS-targeted therapies. SIGNIFICANCE YAP1/TAZ-TEAD activation compensates for loss of KRAS effector signaling, establishing a mechanistic basis for concurrent inhibition of TEAD to enhance the efficacy of KRASG12C-selective inhibitor treatment of KRASG12C-mutant cancers. See related commentary by Johnson and Haigis, p. 4005.
Collapse
Affiliation(s)
- A. Cole Edwards
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Clint A. Stalnecker
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Alexis Jean Morales
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Khalilah E. Taylor
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jennifer E. Klomp
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jeffrey A. Klomp
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Andrew M. Waters
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | - Jill Hallin
- Mirati Therapeutics, Inc., San Diego, California
| | | | - Peter Olson
- Mirati Therapeutics, Inc., San Diego, California
| | - Leonard Post
- Vivace Therapeutics, Inc., San Mateo, California
| | | | - Adrienne D. Cox
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Channing J. Der
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
31
|
Fnaiche A, Chan HC, Paquin A, González Suárez N, Vu V, Li F, Allali-Hassani A, Cao MA, Szewczyk MM, Bolotokova A, Allemand F, Gelin M, Barsyte-Lovejoy D, Santhakumar V, Vedadi M, Guichou JF, Annabi B, Gagnon A. Development of HC-258, a Covalent Acrylamide TEAD Inhibitor That Reduces Gene Expression and Cell Migration. ACS Med Chem Lett 2023; 14:1746-1753. [PMID: 38116405 PMCID: PMC10726447 DOI: 10.1021/acsmedchemlett.3c00386] [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: 09/06/2023] [Revised: 11/22/2023] [Accepted: 11/22/2023] [Indexed: 12/21/2023] Open
Abstract
The transcription factor YAP-TEAD is the downstream effector of the Hippo pathway which controls cell proliferation, apoptosis, tissue repair, and organ growth. Dysregulation of the Hippo pathway has been correlated with carcinogenic processes. A co-crystal structure of TEAD with its endogenous ligand palmitic acid (PA) as well as with flufenamic acid (FA) has been disclosed. Here we report the development of HC-258, which derives from FA and possesses an oxopentyl chain that mimics a molecule of PA as well as an acrylamide that reacts covalently with TEAD's cysteine. HC-258 reduces the CTGF, CYR61, AXL, and NF2 transcript levels and inhibits the migration of MDA-MB-231 breast cancer cells. Co-crystallization with hTEAD2 confirmed that HC-258 binds within TEAD's PA pocket, where it forms a covalent bond with its cysteine.
Collapse
Affiliation(s)
- Ahmed Fnaiche
- Département
de Chimie, Université du Québec
à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Québec H3C 3P8, Canada
| | - Hwai-Chien Chan
- Département
de Chimie, Université du Québec
à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Québec H3C 3P8, Canada
| | - Alexis Paquin
- Département
de Chimie, Université du Québec
à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Québec H3C 3P8, Canada
| | - Narjara González Suárez
- Département
de Chimie, Université du Québec
à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Québec H3C 3P8, Canada
| | - Victoria Vu
- Structural
Genomics Consortium, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Fengling Li
- Structural
Genomics Consortium, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | | | - Michelle Ada Cao
- Structural
Genomics Consortium, 101 College Street, Toronto, Ontario M5G 1L7, Canada
- Department
of Pharmacology and Toxicology, University
of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Magdalena M. Szewczyk
- Structural
Genomics Consortium, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Albina Bolotokova
- Structural
Genomics Consortium, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Frédéric Allemand
- Centre
de Biologie Structurale, CNRS, INSERM, Univ.
Montpellier, 34090 Montpellier, France
| | - Muriel Gelin
- Centre
de Biologie Structurale, CNRS, INSERM, Univ.
Montpellier, 34090 Montpellier, France
| | - Dalia Barsyte-Lovejoy
- Structural
Genomics Consortium, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | | | - Masoud Vedadi
- Department
of Pharmacology and Toxicology, University
of Toronto, Toronto, Ontario M5S 1A8, Canada
- Drug
Discovery Program, Ontario Institute for
Cancer Research, Toronto, Ontario M5G 0A3, Canada
| | - Jean-François Guichou
- Centre
de Biologie Structurale, CNRS, INSERM, Univ.
Montpellier, 34090 Montpellier, France
| | - Borhane Annabi
- Département
de Chimie, Université du Québec
à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Québec H3C 3P8, Canada
| | - Alexandre Gagnon
- Département
de Chimie, Université du Québec
à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Québec H3C 3P8, Canada
| |
Collapse
|
32
|
Evsen L, Morris PJ, Thomas CJ, Ceribelli M. Comparative Assessment and High-Throughput Drug-Combination Profiling of TEAD-Palmitoylation Inhibitors in Hippo Pathway Deficient Mesothelioma. Pharmaceuticals (Basel) 2023; 16:1635. [PMID: 38139762 PMCID: PMC10747288 DOI: 10.3390/ph16121635] [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: 09/29/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 12/24/2023] Open
Abstract
The hippo signaling pathway is a central tumor suppressor cascade frequently inactivated in selected human cancers, leading to the aberrant activation of TEAD transcription factors. Whereas several TEAD auto-palmitoylation inhibitors are currently in development, a comprehensive assessment of this novel drug-modality is missing. Here, we report a comparative analysis among six TEADi(s) using cell-based and biochemical assays in Hippo pathway deficient mesothelioma. Our analysis revealed varying potency and selectivity across TEADi, also highlighting their limited efficacy. To overcome this limitation, we performed an unbiased, quantitative high-throughput drug screening by combining the TEADi VT-103 with a library of approximately 3000 oncology-focused drugs. By exploiting this library's mechanistic redundancy, we identified several drug-classes robustly synergized with TEADi. These included glucocorticoid-receptor (GR) agonists, Mek1/2 inhibitors, mTOR inhibitors, and PI3K inhibitors, among others. Altogether, we report a coherent single-agent dataset informing on potency and selectivity of TEAD-palmitoylation inhibitors as single-agents. We also describe a rational pipeline enabling the systematic identification of TEAD druggable co-dependencies. This data should support the pre-clinical development of drug combination strategies for the treatment of Hippo-deficient mesothelioma, and more broadly, for other cancers dependent on the oncogenic activity of YAP/TEAD.
Collapse
Affiliation(s)
| | | | | | - Michele Ceribelli
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA
| |
Collapse
|
33
|
Shi H, Zou Y, Zhong W, Li Z, Wang X, Yin Y, Li D, Liu Y, Li M. Complex roles of Hippo-YAP/TAZ signaling in hepatocellular carcinoma. J Cancer Res Clin Oncol 2023; 149:15311-15322. [PMID: 37608027 DOI: 10.1007/s00432-023-05272-2] [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: 06/29/2023] [Accepted: 08/09/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND The Hippo signaling pathway is an evolutionarily conserved signaling module that controls organ size in different species, and the disorder of the Hippo pathway can induce liver cancer in organisms, especially hepatocellular carcinoma (HCC). The exact mechanism that causes cancer is still unknown. Recent studies have shown that it is a classical kinase cascade that phosphorylates the Mst1/2-sav1 complex and activates the phosphorylation of the Lats1/2-mob1A/B complex for inactivating Yap and Taz. These kinases and scaffolds are regarded as primary regulators of the Hippo pathway, and help in activating a variety of carcinogenic processes. Among them, Yap/Taz is seen to be the main effector molecule, which is downstream of the Hippo pathway, and its abnormal activation is related to a variety of human cancers including liver cancer. Currently, since Yap/Taz plays a variety of roles in cancer promotion and tumor regeneration, the Hippo pathway has emerged as an attractive target in recent drug development research. METHODS We collect and review relevant literature in web of Science and Pubmed. CONCLUSION This review highlights the important roles of Yap/Taz in activating Hippo pathway in liver cancer. The recent findings on the crosstalks between the Hippo and other cancer associated pathways and moleculars are also discussed. In this review, we summarized and discussed recent breakthroughs in our understanding of how key components of the Hippo-YAP/TAZ pathway influence the hepatocellular carcinoma, including their effects on tumor occurrence and development, their roles in regulating metastasis, and their function in chemotherapy resistance. Further, the molecular mechanism and roles in regulating cross talk between Hippo-YAP/TAZ pathway and other cancer-associated pathways or oncogenes/cancer suppressor genes were summarized and discussed. More, many other inducers and inhibitors of this signaling cascade and available experimental therapies against the YAP/TAZ/TEAD axis were discussed. Targeting this pathway for cancer therapy may have great significance in the treatment of hepatocellular carcinoma. Graphical summary of the complex role of Hippo-YAP/TAZ signaling in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Hewen Shi
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Ying Zou
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Weiwei Zhong
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Zhaoying Li
- Traditional Chinese Medicine Research Center, Shandong Public Health Clinical Center, Jinan, 250102, People's Republic of China
| | - Xiaoxue Wang
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Yancun Yin
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Defang Li
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Ying Liu
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China.
| | - Minjing Li
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China.
| |
Collapse
|
34
|
Shen X, Li Q, Sun Y, Chen L, Xue F, Tian W, Wang Y. The Hippo pathway in endometrial cancer: a potential therapeutic target? Front Oncol 2023; 13:1273345. [PMID: 37927473 PMCID: PMC10625429 DOI: 10.3389/fonc.2023.1273345] [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: 08/06/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
Endometrial cancer, one of the most prevalent malignant cancers tumors of the female reproductive tract, has been increasing in incidence and mortality rates around the world. The Hippo pathway, one of the eight traditional human cancer signaling pathways, is an intricate signaling network that regulates cell proliferation, differentiation, and migration as well as restricting organ size in response to a range of intracellular and extracellular signals. Inhibiting the Hippo pathway results in aberrant activation of its downstream core component YAP/TAZ, which can enhance cancer cells' metabolism and maintain their stemness. Additionally, the Hippo pathway can modulate the tumor microenvironment and induce drug resistance, where tumorigenesis and tumor progression occur. However, the Hippo pathway has been little researched in endometrial cancer. Here, we aim to review how the Hippo pathway contributes to the onset, development and the potential treatment of endometrial cancer with the aim of providing new therapeutic targets.
Collapse
Affiliation(s)
- Xinyun Shen
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Qianqian Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yiqing Sun
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Lingli Chen
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenyan Tian
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
35
|
Fnaiche A, Mélin L, Suárez NG, Paquin A, Vu V, Li F, Allali-Hassani A, Bolotokova A, Allemand F, Gelin M, Cotelle P, Woo S, LaPlante SR, Barsyte-Lovejoy D, Santhakumar V, Vedadi M, Guichou JF, Annabi B, Gagnon A. Development of LM-41 and AF-2112, two flufenamic acid-derived TEAD inhibitors obtained through the replacement of the trifluoromethyl group by aryl rings. Bioorg Med Chem Lett 2023; 95:129488. [PMID: 37770003 DOI: 10.1016/j.bmcl.2023.129488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
The Hippo pathway regulates organ size and tissue homeostasis by controlling cell proliferation and apoptosis. The YAP-TEAD transcription factor, the downstream effector of the Hippo pathway, regulates the expression of genes such as CTGF, Cyr61, Axl and NF2. Aberrant Hippo activity has been identified in multiple types of cancers. Flufenamic acid (FA) was reported to bind in a liphophilic TEAD palmitic acid (PA) pocket, leading to reduction of the expression of Axl and NF2. Here, we show that the replacement of the trifluoromethyl moiety in FA by aromatic groups, directly connected to the scaffold or separated by a linker, leads to compounds with better affinity to TEAD. Co-crystallization studies show that these compounds bind similarly to FA, but deeper within the PA pocket. Our studies identified LM-41 and AF-2112 as two TEAD binders that strongly reduce the expression of CTGF, Cyr61, Axl and NF2. LM-41 gave the strongest reduction of migration of human MDA-MB-231 breast cancer cells.
Collapse
Affiliation(s)
- Ahmed Fnaiche
- Département de chimie, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Québec H3C 3P8, Canada
| | - Léa Mélin
- Département de chimie, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Québec H3C 3P8, Canada
| | - Narjara González Suárez
- Département de chimie, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Québec H3C 3P8, Canada
| | - Alexis Paquin
- Département de chimie, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Québec H3C 3P8, Canada
| | - Victoria Vu
- Structural Genomics Consortium, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Fengling Li
- Structural Genomics Consortium, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | | | - Albina Bolotokova
- Structural Genomics Consortium, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Frédéric Allemand
- Centre de Biologie Structurale, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Muriel Gelin
- Centre de Biologie Structurale, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Philippe Cotelle
- Université de Lille, CHU Lille, INSERM-UMR-S-1172-JPArc-Centre de Recherche Jean-Pierre Aubert, Neurosciences et Cancer, F-59000 Lille, France
| | - Simon Woo
- INRS-Centre Armand Frappier Santé Biotechnologie, Université du Québec, 531 Boulevard des Prairies, Laval, Québec H7V 1B7, Canada
| | - Steven R LaPlante
- INRS-Centre Armand Frappier Santé Biotechnologie, Université du Québec, 531 Boulevard des Prairies, Laval, Québec H7V 1B7, Canada
| | - Dalia Barsyte-Lovejoy
- Structural Genomics Consortium, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | | | - Masoud Vedadi
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Jean-François Guichou
- Centre de Biologie Structurale, CNRS, INSERM, Univ. Montpellier, Montpellier, France.
| | - Borhane Annabi
- Département de chimie, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Québec H3C 3P8, Canada.
| | - Alexandre Gagnon
- Département de chimie, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Québec H3C 3P8, Canada.
| |
Collapse
|
36
|
Kofler M, Kapus A. Nuclear Import and Export of YAP and TAZ. Cancers (Basel) 2023; 15:4956. [PMID: 37894323 PMCID: PMC10605228 DOI: 10.3390/cancers15204956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/05/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Yes-associated Protein (YAP) and its paralog Transcriptional Coactivator with PDZ-binding Motif (TAZ) are major regulators of gene transcription/expression, primarily controlled by the Hippo pathway and the cytoskeleton. Integrating an array of chemical and mechanical signals, they impact growth, differentiation, and regeneration. Accordingly, they also play key roles in tumorigenesis and metastasis formation. Their activity is primarily regulated by their localization, that is, Hippo pathway- and/or cytoskeleton-controlled cytosolic or nuclear sequestration. While many details of such prevailing retention models have been elucidated, much less is known about their actual nuclear traffic: import and export. Although their size is not far from the cutoff for passive diffusion through the nuclear pore complex (NPC), and they do not contain any classic nuclear localization (NLS) or nuclear export signal (NES), evidence has been accumulating that their shuttling involves mediated and thus regulatable/targetable processes. The aim of this review is to summarize emerging information/concepts about their nucleocytoplasmic shuttling, encompassing the relevant structural requirements (NLS, NES), nuclear transport receptors (NTRs, karyophererins), and NPC components, along with the potential transport mechanisms and their regulation. While dissecting retention vs. transport is often challenging, the emerging picture suggests that YAP/TAZ shuttles across the NPC via multiple, non-exclusive, mediated mechanisms, constituting a novel and intriguing facet of YAP/TAZ biology.
Collapse
Affiliation(s)
- Michael Kofler
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada;
| | - András Kapus
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada;
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
37
|
Sabnis RW. Novel Indazole Compounds as TEAD Inhibitors for Treating Cancer. ACS Med Chem Lett 2023; 14:1334-1335. [PMID: 37849539 PMCID: PMC10577882 DOI: 10.1021/acsmedchemlett.3c00392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Indexed: 10/19/2023] Open
Abstract
Provided herein are novel indazole compounds as TEAD inhibitors, pharmaceutical compositions, use of such compounds in treating cancer, and processes for preparing such compounds.
Collapse
Affiliation(s)
- Ram W. Sabnis
- Smith, Gambrell & Russell
LLP, 1105 W. Peachtree Street NE, Suite 1000, Atlanta, Georgia 30309, United States
| |
Collapse
|
38
|
El Yousfi Y, Mora-Molina R, López-Rivas A, Yerbes R. Role of the YAP/TAZ-TEAD Transcriptional Complex in the Metabolic Control of TRAIL Sensitivity by the Mevalonate Pathway in Cancer Cells. Cells 2023; 12:2370. [PMID: 37830584 PMCID: PMC10571597 DOI: 10.3390/cells12192370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/14/2023] Open
Abstract
Different studies have reported that inhibiting the mevalonate pathway with statins may increase the sensitivity of cancer cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), although the signaling mechanism leading to this sensitization remains largely unknown. We investigated the role of the YAP (Yes-associated protein)/TAZ (transcriptional co-activator with PDZ-binding motif)-TEAD (TEA/ATTS domain) transcriptional complex in the metabolic control of TRAIL sensitivity by the mevalonate pathway. We show that depleting nuclear YAP/TAZ in tumor cells, either via treatment with statins or by silencing YAP/TAZ expression with siRNAs, facilitates the activation of apoptosis by TRAIL. Furthermore, the blockage of TEAD transcriptional activity either pharmacologically or through the ectopic expression of a disruptor of the YAP/TAZ interaction with TEAD transcription factors, overcomes the resistance of tumor cells to the induction of apoptosis by TRAIL. Our results show that the mevalonate pathway controls cellular the FLICE-inhibitory protein (cFLIP) expression in tumor cells. Importantly, inhibiting the YAP/TAZ-TEAD signaling pathway induces cFLIP down-regulation, leading to a marked sensitization of tumor cells to apoptosis induction by TRAIL. Our data suggest that a combined strategy of targeting TEAD activity and selectively activating apoptosis signaling by agonists of apoptotic TRAIL receptors could be explored as a potential therapeutic approach in cancer treatment.
Collapse
Affiliation(s)
- Younes El Yousfi
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, 41092 Seville, Spain; (Y.E.Y.); (R.M.-M.); (A.L.-R.)
| | - Rocío Mora-Molina
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, 41092 Seville, Spain; (Y.E.Y.); (R.M.-M.); (A.L.-R.)
| | - Abelardo López-Rivas
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, 41092 Seville, Spain; (Y.E.Y.); (R.M.-M.); (A.L.-R.)
| | - Rosario Yerbes
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, 41092 Seville, Spain; (Y.E.Y.); (R.M.-M.); (A.L.-R.)
- Medical Physiology and Biophysics Department, Universidad de Sevilla and Instituto de Biomedicina de Sevilla (IBiS) (Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla), 41013 Seville, Spain
| |
Collapse
|
39
|
Sabnis RW. Novel Compounds as TEAD Inhibitors for Treating Cancer. ACS Med Chem Lett 2023; 14:1152-1153. [PMID: 37736181 PMCID: PMC10510495 DOI: 10.1021/acsmedchemlett.3c00346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Indexed: 09/23/2023] Open
Abstract
Provided herein are novel compounds as TEAD inhibitors, pharmaceutical compositions, use of such compounds in treating cancer, and processes for preparing such compounds.
Collapse
Affiliation(s)
- Ram W. Sabnis
- Smith, Gambrell & Russell LLP, 1105 W. Peachtree Street NE, Suite
1000, Atlanta, Georgia 30309, United States
| |
Collapse
|
40
|
Benton D, Chernoff J. TRIMming away colon cancer: TRIM21-mediated ubiquitination as an activator of the Hippo tumor suppressor pathway. Cell Chem Biol 2023; 30:699-701. [PMID: 37478825 DOI: 10.1016/j.chembiol.2023.06.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/23/2023]
Abstract
In this issue of Cell Chemical Biology, Liu et al.1 identify TRIM21-mediated ubiquitination of the Hippo pathway kinase MST2, promoting its dimerization and activation. The antidepressant Vilazodone was found to bind to TRIM21, enhancing its activity toward MST2, increasing Hippo activation, and reducing colorectal cancer metastasis.
Collapse
Affiliation(s)
- Dorothy Benton
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Jonathan Chernoff
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA.
| |
Collapse
|
41
|
Pontes B, Mendes FA. Mechanical Properties of Glioblastoma: Perspectives for YAP/TAZ Signaling Pathway and Beyond. Diseases 2023; 11:86. [PMID: 37366874 DOI: 10.3390/diseases11020086] [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: 05/10/2023] [Revised: 06/06/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023] Open
Abstract
Glioblastoma is a highly aggressive brain tumor with a poor prognosis. Recent studies have suggested that mechanobiology, the study of how physical forces influence cellular behavior, plays an important role in glioblastoma progression. Several signaling pathways, molecules, and effectors, such as focal adhesions, stretch-activated ion channels, or membrane tension variations, have been studied in this regard. Also investigated are YAP/TAZ, downstream effectors of the Hippo pathway, which is a key regulator of cell proliferation and differentiation. In glioblastoma, YAP/TAZ have been shown to promote tumor growth and invasion by regulating genes involved in cell adhesion, migration, and extracellular matrix remodeling. YAP/TAZ can be activated by mechanical cues such as cell stiffness, matrix rigidity, and cell shape changes, which are all altered in the tumor microenvironment. Furthermore, YAP/TAZ have been shown to crosstalk with other signaling pathways, such as AKT, mTOR, and WNT, which are dysregulated in glioblastoma. Thus, understanding the role of mechanobiology and YAP/TAZ in glioblastoma progression could provide new insights into the development of novel therapeutic strategies. Targeting YAP/TAZ and mechanotransduction pathways in glioblastoma may offer a promising approach to treating this deadly disease.
Collapse
Affiliation(s)
- Bruno Pontes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Fabio A Mendes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| |
Collapse
|
42
|
Habshi T, Shelke V, Kale A, Lech M, Bhanudas Gaikwad A. Hippo signaling in acute kidney injury to chronic kidney disease transition: current understandings and future targets. Drug Discov Today 2023:103649. [PMID: 37268185 DOI: 10.1016/j.drudis.2023.103649] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/19/2023] [Accepted: 05/26/2023] [Indexed: 06/04/2023]
Abstract
Acute kidney injury (AKI)-to-chronic kidney disease (CKD) transition is a slow but persistent progression toward end-stage kidney disease. Earlier reports have shown that Hippo components, such as Yes-associated protein (YAP) and its homolog TAZ (Transcriptional coactivator with PDZ-binding motif), regulate inflammation and fibrogenesis during the AKI-to-CKD transition. Notably, the roles and mechanisms of Hippo components vary during AKI, AKI-to-CKD transition, and CKD. Hence, it is important to understand these roles in detail. This review addresses the potential of Hippo regulators or components as future therapeutic targets for halting the AKI-to-CKD transition.
Collapse
Affiliation(s)
- Tahib Habshi
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan-333031, India
| | - Vishwadeep Shelke
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan-333031, India
| | - Ajinath Kale
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan-333031, India
| | - Maciej Lech
- Division of Nephrology, Department of Internal Medicine IV, Hospital of the Ludwig Maximilians University Munich, 80336 Munich, Germany
| | - Anil Bhanudas Gaikwad
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan-333031, India.
| |
Collapse
|
43
|
Neil E, Kouskoff V. Current Model Systems for Investigating Epithelioid Haemangioendothelioma. Cancers (Basel) 2023; 15:3005. [PMID: 37296967 PMCID: PMC10251951 DOI: 10.3390/cancers15113005] [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: 04/12/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Epithelioid haemangioendothelioma (EHE) is a rare sarcoma of the vascular endothelium with an unpredictable disease course. EHE tumours can remain indolent for long period of time but may suddenly evolve into an aggressive disease with widespread metastases and a poor prognosis. Two mutually exclusive chromosomal translocations define EHE tumours, each involving one of the transcription co-factors TAZ and YAP. The TAZ-CAMTA1 fusion protein results from a t(1;3) translocation and is present in 90% of EHE tumours. The remaining 10% of EHE cases harbour a t(X;11) translocation, resulting in the YAP1-TFE3 (YT) fusion protein. Until recently, the lack of representative EHE models made it challenging to study the mechanisms by which these fusion proteins promote tumorigenesis. Here, we describe and compare the recently developed experimental approaches that are currently available for studying this cancer. After summarising the key findings obtained with each experimental approach, we discuss the advantages and limitations of these different model systems. Our survey of the current literature shows how each experimental approach can be utilised in different ways to improve our understanding of EHE initiation and progression. Ultimately, this should lead to better treatment options for patients.
Collapse
Affiliation(s)
- Emily Neil
- School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Valerie Kouskoff
- School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
44
|
Luo J, Deng L, Zou H, Guo Y, Tong T, Huang M, Ling G, Li P. New insights into the ambivalent role of YAP/TAZ in human cancers. J Exp Clin Cancer Res 2023; 42:130. [PMID: 37211598 DOI: 10.1186/s13046-023-02704-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/10/2023] [Indexed: 05/23/2023] Open
Abstract
Hippo signaling was first identified in Drosophila as a key controller of organ size by regulating cell proliferation and anti-apoptosis. Subsequent studies have shown that this pathway is highly conserved in mammals, and its dysregulation is implicated in multiple events of cancer development and progression. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) (hereafter YAP/TAZ) are the downstream effectors of the Hippo pathway. YAP/TAZ overexpression or activation is sufficient to induce tumor initiation and progression, as well as recurrence and therapeutic resistance. However, there is growing evidence that YAP/TAZ also exert a tumor-suppressive function in a context-dependent manner. Therefore, caution should be taken when targeting Hippo signaling in clinical trials in the future. In this review article, we will first give an overview of YAP/TAZ and their oncogenic roles in various cancers and then systematically summarize the tumor-suppressive functions of YAP/TAZ in different contexts. Based on these findings, we will further discuss the clinical implications of YAP/TAZ-based tumor targeted therapy and potential future directions.
Collapse
Affiliation(s)
- Juan Luo
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, People's Republic of China
| | - Liang Deng
- Department of General Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, People's Republic of China
| | - Hailin Zou
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, People's Republic of China
| | - Yibo Guo
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, People's Republic of China
| | - Tongyu Tong
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, People's Republic of China
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, People's Republic of China
| | - Mingli Huang
- Department of General Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, People's Republic of China
| | - Gengqiang Ling
- Department of Neurosurgery, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, People's Republic of China
| | - Peng Li
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, People's Republic of China.
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, People's Republic of China.
| |
Collapse
|
45
|
Zhao B, Pobbati AV, Rubin BP, Stauffer S. Leveraging Hot Spots of TEAD-Coregulator Interactions in the Design of Direct Small Molecule Protein-Protein Interaction Disruptors Targeting Hippo Pathway Signaling. Pharmaceuticals (Basel) 2023; 16:ph16040583. [PMID: 37111340 PMCID: PMC10146773 DOI: 10.3390/ph16040583] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
The Hippo signaling pathway is a highly conserved pathway that plays important roles in the regulation of cell proliferation and apoptosis. Transcription factors TEAD1-4 and transcriptional coregulators YAP/TAZ are the downstream effectors of the Hippo pathway and can modulate Hippo biology. Dysregulation of this pathway is implicated in tumorigenesis and acquired resistance to therapies. The emerging importance of YAP/TAZ-TEAD interaction in cancer development makes it a potential therapeutic target. In the past decade, disrupting YAP/TAZ-TEAD interaction as an effective approach for cancer treatment has achieved great progress. This approach followed a trajectory wherein peptidomimetic YAP-TEAD protein-protein interaction disruptors (PPIDs) were first designed, followed by the discovery of allosteric small molecule PPIDs, and currently, the development of direct small molecule PPIDs. YAP and TEAD form three interaction interfaces. Interfaces 2 and 3 are amenable for direct PPID design. One direct YAP-TEAD PPID (IAG933) that targets interface 3 has entered a clinical trial in 2021. However, in general, strategically designing effective small molecules PPIDs targeting TEAD interfaces 2 and 3 has been challenging compared with allosteric inhibitor development. This review focuses on the development of direct surface disruptors and discusses the challenges and opportunities for developing potent YAP/TAZ-TEAD inhibitors for the treatment of cancer.
Collapse
Affiliation(s)
- Bin Zhao
- Cleveland Clinic Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44106, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Ajaybabu V Pobbati
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Brian P Rubin
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Shaun Stauffer
- Cleveland Clinic Center for Therapeutics Discovery, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44106, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| |
Collapse
|