1
|
Blackwell JH, Michaelides IN, Gibault F. A Perspective on the Strategic Application of Deconstruction-Reconstruction in Drug Discovery. J Med Chem 2025. [PMID: 40324045 DOI: 10.1021/acs.jmedchem.5c00036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
The deconstruction of known ligands to proteins into smaller, often fragment-sized components and subsequent regrowth from these leaner starting points has been shown to have interesting and useful application in medicinal chemistry. Such efforts can provide excellent start points for renewed hit-to-lead campaigns while assisting medicinal chemists in gaining a deeper understanding on the ligandability of a protein. Selected case-studies making strategic use of the deconstruction-reconstruction logic are examined herein, thus aiming to highlight the value of employing this approach in drug discovery programs.
Collapse
Affiliation(s)
- J Henry Blackwell
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, United Kingdom
| | - Iacovos N Michaelides
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, United Kingdom
| | - Floriane Gibault
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, United Kingdom
| |
Collapse
|
2
|
Bao Q, Kumar A, Wu D, Zhou J. Targeting EED as a key PRC2 complex mediator toward novel epigenetic therapeutics. Drug Discov Today 2024; 29:103986. [PMID: 38642703 PMCID: PMC11416859 DOI: 10.1016/j.drudis.2024.103986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/06/2024] [Accepted: 04/15/2024] [Indexed: 04/22/2024]
Abstract
EED within the PRC2 complex is crucial for chromatin regulation particularly in tumor development, making its inhibition a promising epigenetic therapeutic strategy. Significant advancement in PRC2 inhibitor development has been achieved with an approved EZH2 inhibitor in the market and with others in the clinical trials. However, current EZH2 inhibitors are limited to specific blood cancers and encounter therapeutic resistance. EED stabilizes PRC2 complex and enhances its activity through unique allosteric mechanisms, thereby acting as both a scaffold protein and a recognizer of H3K27me3 making it an attractive drug target. This review provides an overview of epigenetic therapeutic strategies targeting EED, including allosteric inhibitors, PPI inhibitors, and PROTACs, together with brief discussions on the relevant challenges, opportunities, and future directions.
Collapse
Affiliation(s)
- Qichao Bao
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Anil Kumar
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Daqing Wu
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA 30314, USA
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
3
|
Gao M, Li Y, Cao P, Liu H, Chen J, Kang S. Exploring the therapeutic potential of targeting polycomb repressive complex 2 in lung cancer. Front Oncol 2023; 13:1216289. [PMID: 37909018 PMCID: PMC10613995 DOI: 10.3389/fonc.2023.1216289] [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: 05/04/2023] [Accepted: 10/02/2023] [Indexed: 11/02/2023] Open
Abstract
The pathogenesis of lung cancer (LC) is a multifaceted process that is influenced by a variety of factors. Alongside genetic mutations and environmental influences, there is increasing evidence that epigenetic mechanisms play a significant role in the development and progression of LC. The Polycomb repressive complex 2 (PRC2), composed of EZH1/2, SUZ12, and EED, is an epigenetic silencer that controls the expression of target genes and is crucial for cell identity in multicellular organisms. Abnormal expression of PRC2 has been shown to contribute to the progression of LC through several pathways. Although targeted inhibition of EZH2 has demonstrated potential in delaying the progression of LC and improving chemotherapy sensitivity, the effectiveness of enzymatic inhibitors of PRC2 in LC is limited, and a more comprehensive understanding of PRC2's role is necessary. This paper reviews the core subunits of PRC2 and their interactions, and outlines the mechanisms of aberrant PRC2 expression in cancer and its role in tumor immunity. We also summarize the important role of PRC2 in regulating biological behaviors such as epithelial mesenchymal transition, invasive metastasis, apoptosis, cell cycle regulation, autophagy, and PRC2-mediated resistance to LC chemotherapeutic agents in LC cells. Lastly, we explored the latest breakthroughs in the research and evaluation of medications that target PRC2, as well as the latest findings from clinical studies investigating the efficacy of these drugs in the treatment of various human cancers.
Collapse
Affiliation(s)
- Min Gao
- Department of Thoracic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Inner Mongolia Medical University, First Clinical Medical College, Hohhot, China
| | - Yongwen Li
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Peijun Cao
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongyu Liu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jun Chen
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Shirong Kang
- Department of Thoracic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
4
|
Li X, Chen Y, Bai L, Zhao R, Wu Y, Xie ZR, Wu JM, Bowen NJ, Danaher A, Cook N, Li D, Qui M, Du Y, Fu H, Osunkoya AO, Kucuk O, Wu D. Nicardipine is a putative EED inhibitor and has high selectivity and potency against chemoresistant prostate cancer in preclinical models. Br J Cancer 2023; 129:884-894. [PMID: 37474721 PMCID: PMC10449793 DOI: 10.1038/s41416-023-02359-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 06/22/2023] [Accepted: 06/29/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND It is imperative to develop novel therapeutics to overcome chemoresistance, a significant obstacle in the clinical management of prostate cancer (PCa) and other cancers. METHODS A phenotypic screen was performed to identify novel inhibitors of chemoresistant PCa cells. The mechanism of action of potential candidate(s) was investigated using in silico docking, and molecular and cellular assays in chemoresistant PCa cells. The in vivo efficacy was evaluated in mouse xenograft models of chemoresistant PCa. RESULTS Nicardipine exhibited high selectivity and potency against chemoresistant PCa cells via inducing apoptosis and cell cycle arrest. Computational, molecular, and cellular studies identified nicardipine as a putative inhibitor of embryonic ectoderm development (EED) protein, and the results are consistent with a proposed mechanism of action that nicardipine destabilised enhancer of zeste homologue 2 (EZH2) and inhibited key components of noncanonical EZH2 signalling, including transducer and activator of transcription 3, S-phase kinase-associated protein 2, ATP binding cassette B1, and survivin. As a monotherapy, nicardipine effectively inhibited the skeletal growth of chemoresistant C4-2B-TaxR tumours. As a combination regimen, nicardipine synergistically enhanced the in vivo efficacy of docetaxel against C4-2 xenografts. CONCLUSION Our findings provided the first preclinical evidence supporting nicardipine as a novel EED inhibitor that has the potential to be promptly tested in PCa patients to overcome chemoresistance and improve clinical outcomes.
Collapse
Affiliation(s)
- Xin Li
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, USA
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Yanhua Chen
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lijuan Bai
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Rui Zhao
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Yifei Wu
- School of Electrical and Computer Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - Zhong-Ru Xie
- School of Electrical and Computer Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - Jason M Wu
- Emory College of Arts and Sciences, Atlanta, GA, USA
| | - Nathan J Bowen
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, USA
| | - Alira Danaher
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, USA
| | - Nicholas Cook
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, USA
| | - Dehong Li
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, USA
| | - Min Qui
- Department of Pharmacology and Chemical Biology, and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Yuhong Du
- Department of Pharmacology and Chemical Biology, and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Haian Fu
- Department of Pharmacology and Chemical Biology, and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Adeboye O Osunkoya
- Departments of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Omer Kucuk
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Daqing Wu
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, USA.
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA.
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA.
- MetCure Therapeutics LLC, Atlanta, GA, USA.
| |
Collapse
|
5
|
Vulpetti A, Holzer P, Schmiedeberg N, Imbach-Weese P, Pissot-Soldermann C, Hollingworth GJ, Radimerski T, Thoma CR, Stachyra TM, Wojtynek M, Maschlej M, Chau S, Schuffenhauer A, Fernández C, Schröder M, Renatus M. Discovery of New Binders for DCAF1, an Emerging Ligase Target in the Targeted Protein Degradation Field. ACS Med Chem Lett 2023; 14:949-954. [PMID: 37465299 PMCID: PMC10350940 DOI: 10.1021/acsmedchemlett.3c00104] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/31/2023] [Indexed: 07/20/2023] Open
Abstract
In this study, we describe the rapid identification of potent binders for the WD40 repeat domain (WDR) of DCAF1. This was achieved by two rounds of iterative focused screening of a small set of compounds selected on the basis of internal WDR domain knowledge followed by hit expansion. Subsequent structure-based design led to nanomolar potency binders with a clear exit vector enabling DCAF1-based bifunctional degrader exploration.
Collapse
Affiliation(s)
- Anna Vulpetti
- Global
Discovery Chemistry, Novartis Institutes
for BioMedical Research, Basel 4002, Switzerland
| | - Philipp Holzer
- Global
Discovery Chemistry, Novartis Institutes
for BioMedical Research, Basel 4002, Switzerland
| | - Niko Schmiedeberg
- Global
Discovery Chemistry, Novartis Institutes
for BioMedical Research, Basel 4002, Switzerland
| | - Patricia Imbach-Weese
- Global
Discovery Chemistry, Novartis Institutes
for BioMedical Research, Basel 4002, Switzerland
| | - Carole Pissot-Soldermann
- Global
Discovery Chemistry, Novartis Institutes
for BioMedical Research, Basel 4002, Switzerland
| | - Gregory J. Hollingworth
- Global
Discovery Chemistry, Novartis Institutes
for BioMedical Research, Basel 4002, Switzerland
| | - Thomas Radimerski
- Oncology
Drug Discovery, Novartis Institutes for
BioMedical Research, Basel 4002, Switzerland
| | - Claudio R. Thoma
- Oncology
Drug Discovery, Novartis Institutes for
BioMedical Research, Basel 4002, Switzerland
| | - Therese-Marie Stachyra
- Oncology
Drug Discovery, Novartis Institutes for
BioMedical Research, Basel 4002, Switzerland
| | - Matthias Wojtynek
- Chemical
Biology & Therapeutics, Novartis Institutes
for BioMedical Research, Basel 4002, Switzerland
| | - Magdalena Maschlej
- Chemical
Biology & Therapeutics, Novartis Institutes
for BioMedical Research, Basel 4002, Switzerland
| | - Suzanne Chau
- Chemical
Biology & Therapeutics, Novartis Institutes
for BioMedical Research, Basel 4002, Switzerland
| | - Ansgar Schuffenhauer
- Chemical
Biology & Therapeutics, Novartis Institutes
for BioMedical Research, Basel 4002, Switzerland
| | - César Fernández
- Chemical
Biology & Therapeutics, Novartis Institutes
for BioMedical Research, Basel 4002, Switzerland
| | - Martin Schröder
- Chemical
Biology & Therapeutics, Novartis Institutes
for BioMedical Research, Basel 4002, Switzerland
| | - Martin Renatus
- Chemical
Biology & Therapeutics, Novartis Institutes
for BioMedical Research, Basel 4002, Switzerland
| |
Collapse
|
6
|
Huang Y, Sendzik M, Zhang J, Gao Z, Sun Y, Wang L, Gu J, Zhao K, Yu Z, Zhang L, Zhang Q, Blanz J, Chen Z, Dubost V, Fang D, Feng L, Fu X, Kiffe M, Li L, Luo F, Luo X, Mi Y, Mistry P, Pearson D, Piaia A, Scheufler C, Terranova R, Weiss A, Zeng J, Zhang H, Zhang J, Zhao M, Dillon MP, Jeay S, Qi W, Moggs J, Pissot-Soldermann C, Li E, Atadja P, Lingel A, Oyang C. Discovery of the Clinical Candidate MAK683: An EED-Directed, Allosteric, and Selective PRC2 Inhibitor for the Treatment of Advanced Malignancies. J Med Chem 2022; 65:5317-5333. [PMID: 35352560 DOI: 10.1021/acs.jmedchem.1c02148] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Polycomb Repressive Complex 2 (PRC2) plays an important role in transcriptional regulation during animal development and in cell differentiation, and alteration of PRC2 activity has been associated with cancer. On a molecular level, PRC2 catalyzes methylation of histone H3 lysine 27 (H3K27), resulting in mono-, di-, or trimethylated forms of H3K27, of which the trimethylated form H3K27me3 leads to transcriptional repression of polycomb target genes. Previously, we have shown that binding of the low-molecular-weight compound EED226 to the H3K27me3 binding pocket of the regulatory subunit EED can effectively inhibit PRC2 activity in cells and reduce tumor growth in mouse xenograft models. Here, we report the stepwise optimization of the tool compound EED226 toward the potent and selective EED inhibitor MAK683 (compound 22) and its subsequent preclinical characterization. Based on a balanced PK/PD profile, efficacy, and mitigated risk of forming reactive metabolites, MAK683 has been selected for clinical development.
Collapse
Affiliation(s)
- Ying Huang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Martin Sendzik
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States.,Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Jeff Zhang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Zhenting Gao
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Yongfeng Sun
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Long Wang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Justin Gu
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Kehao Zhao
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Zhengtian Yu
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Lijun Zhang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Qiong Zhang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Joachim Blanz
- Novartis Institutes for BioMedical Research, Novartis Campus, 4056 Basel, Switzerland
| | - Zijun Chen
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Valérie Dubost
- Novartis Institutes for BioMedical Research, Novartis Campus, 4056 Basel, Switzerland
| | - Douglas Fang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Lijian Feng
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Xingnian Fu
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Michael Kiffe
- Novartis Institutes for BioMedical Research, Novartis Campus, 4056 Basel, Switzerland
| | - Ling Li
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Fangjun Luo
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Xiao Luo
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Yuan Mi
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Prakash Mistry
- Novartis Institutes for BioMedical Research, Novartis Campus, 4056 Basel, Switzerland
| | - David Pearson
- Novartis Institutes for BioMedical Research, Novartis Campus, 4056 Basel, Switzerland
| | - Alessandro Piaia
- Novartis Institutes for BioMedical Research, Novartis Campus, 4056 Basel, Switzerland
| | - Clemens Scheufler
- Novartis Institutes for BioMedical Research, Novartis Campus, 4056 Basel, Switzerland
| | - Remi Terranova
- Novartis Institutes for BioMedical Research, Novartis Campus, 4056 Basel, Switzerland
| | - Andreas Weiss
- Novartis Institutes for BioMedical Research, Novartis Campus, 4056 Basel, Switzerland
| | - Jue Zeng
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Hailong Zhang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Jiangwei Zhang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Mengxi Zhao
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Michael P Dillon
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Sebastien Jeay
- Novartis Institutes for BioMedical Research, Novartis Campus, 4056 Basel, Switzerland
| | - Wei Qi
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Jonathan Moggs
- Novartis Institutes for BioMedical Research, Novartis Campus, 4056 Basel, Switzerland
| | | | - En Li
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Peter Atadja
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Andreas Lingel
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States.,Novartis Institutes for BioMedical Research, Novartis Campus, 4056 Basel, Switzerland
| | - Counde Oyang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| |
Collapse
|
7
|
Del Moral-Morales A, González-Orozco JC, Hernández-Vega AM, Hernández-Ortega K, Peña-Gutiérrez KM, Camacho-Arroyo I. EZH2 Mediates Proliferation, Migration, and Invasion Promoted by Estradiol in Human Glioblastoma Cells. Front Endocrinol (Lausanne) 2022; 13:703733. [PMID: 35197928 PMCID: PMC8859835 DOI: 10.3389/fendo.2022.703733] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
Glioblastomas (GBM) are the most frequent and aggressive brain tumors. 17β-estradiol (E2) increases proliferation, migration, and invasion of human GBM cells; however underlying mechanisms are no fully understood. Zeste 2 Enhancer Homologous enzyme (EZH2) is a methyltransferase part of Polycomb 2 repressor complex (PRC2). In GBM, EZH2 is overexpressed and involved in the cell cycle, migration, and invasion processes. We studied the role of EZH2 in the pro-oncogenic actions of E2 in human GBM cells. EZH2 gene silencing and pharmacological inhibition of EZH2 blocked proliferation, migration, and invasion of GBM cells induced by E2. We identified in silico additional putative estrogen response elements (EREs) at the EZH2 promoter, but E2 did not modify EZH2 expression. In silico analysis also revealed that among human GBM samples, EZH2 expression was homogeneous; in contrast, the heterogeneous expression of estrogen receptors (ERs) allowed the classification of the samples into groups. Even in the GBM cluster with high expression of ERs and those of their target genes, the expression of PCR2 target genes did not change. Overall, our data suggest that in GBM cells, pro-oncogenic actions of E2 are mediated by EZH2, without changes in EZH2 expression and by mechanisms that appear to be unrelated to the transcriptional activity of ERs.
Collapse
Affiliation(s)
- Aylin Del Moral-Morales
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Juan Carlos González-Orozco
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Ana María Hernández-Vega
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Karina Hernández-Ortega
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Karla Mariana Peña-Gutiérrez
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| |
Collapse
|
8
|
Liu KL, Zhu K, Zhang H. An overview of the development of EED inhibitors to disable the PRC2 function. RSC Med Chem 2022; 13:39-53. [PMID: 35224495 PMCID: PMC8792826 DOI: 10.1039/d1md00274k] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 10/21/2021] [Indexed: 10/24/2023] Open
Abstract
Polycomb repressive complex 2 (PRC2) catalyzes the methylation of histone H3 lysine 27 (H3K27) and the enrichment of its catalytic product H3K27me3 is responsible for the silencing of tumor suppressor genes and the blocking of transcripts related to immunity and cell terminal differentiation. Aberrations of PRC2 components, such as mutation and overexpression, have been observed in various cancers, which makes PRC2 a potential therapeutic target for cancer. Up to now, targeting the enhancer of zeste homolog 2 (EZH2), the catalytic subunit of PRC2, represents the main strategy in the development of PRC2 inhibitors. Although significant progress has been made, new problems also emerge, e.g. the drug resistance caused by secondary mutations. In recent years, more and more efforts have shifted to another new strategy - targeting embryonic ectoderm development (EED) to disrupt its major interactions with other components, which are necessary to the PRC2 function, and some promising results have been obtained. This review summarizes the recent development of EED inhibitors as possible chemotherapy for cancer treatment, which could help accelerate future related research work.
Collapse
Affiliation(s)
- Kai-Lu Liu
- School of Biological Science and Technology, University of Jinan Jinan 250022 China
| | - Kongkai Zhu
- School of Biological Science and Technology, University of Jinan Jinan 250022 China
| | - Hua Zhang
- School of Biological Science and Technology, University of Jinan Jinan 250022 China
| |
Collapse
|
9
|
Vijayanathan M, Trejo-Arellano MG, Mozgová I. Polycomb Repressive Complex 2 in Eukaryotes-An Evolutionary Perspective. EPIGENOMES 2022; 6:3. [PMID: 35076495 PMCID: PMC8788455 DOI: 10.3390/epigenomes6010003] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/12/2022] [Accepted: 01/12/2022] [Indexed: 12/23/2022] Open
Abstract
Polycomb repressive complex 2 (PRC2) represents a group of evolutionarily conserved multi-subunit complexes that repress gene transcription by introducing trimethylation of lysine 27 on histone 3 (H3K27me3). PRC2 activity is of key importance for cell identity specification and developmental phase transitions in animals and plants. The composition, biochemistry, and developmental function of PRC2 in animal and flowering plant model species are relatively well described. Recent evidence demonstrates the presence of PRC2 complexes in various eukaryotic supergroups, suggesting conservation of the complex and its function. Here, we provide an overview of the current understanding of PRC2-mediated repression in different representatives of eukaryotic supergroups with a focus on the green lineage. By comparison of PRC2 in different eukaryotes, we highlight the possible common and diverged features suggesting evolutionary implications and outline emerging questions and directions for future research of polycomb repression and its evolution.
Collapse
Affiliation(s)
- Mallika Vijayanathan
- Biology Centre, Institute of Plant Molecular Biology, Czech Academy of Sciences, 370 05 Ceske Budejovice, Czech Republic; (M.V.); (M.G.T.-A.)
| | - María Guadalupe Trejo-Arellano
- Biology Centre, Institute of Plant Molecular Biology, Czech Academy of Sciences, 370 05 Ceske Budejovice, Czech Republic; (M.V.); (M.G.T.-A.)
| | - Iva Mozgová
- Biology Centre, Institute of Plant Molecular Biology, Czech Academy of Sciences, 370 05 Ceske Budejovice, Czech Republic; (M.V.); (M.G.T.-A.)
- Faculty of Science, University of South Bohemia, 370 05 Ceske Budejovice, Czech Republic
| |
Collapse
|
10
|
Tomassi S, Romanelli A, Zwergel C, Valente S, Mai A. Polycomb Repressive Complex 2 Modulation through the Development of EZH2-EED Interaction Inhibitors and EED Binders. J Med Chem 2021; 64:11774-11797. [PMID: 34351144 PMCID: PMC8404197 DOI: 10.1021/acs.jmedchem.1c00226] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
![]()
Epigenetics is nowadays
a well-accepted area of research. In the
last years, tremendous progress was made regarding molecules targeting
EZH2, directly or indirectly. Recently tazemetostat hit the market
after FDA-approval for the treatment of lymphoma. However, the impairment
of EZH2 activity by orthosteric intervention has proven to be effective
only in a limited subset of cancers. Considering the multiproteic
nature of the PRC2 complex and the marked dependence of EZH2 functions
on the other core subunits such as EED, in recent years, a new targeting
approach ascended to prominence. The possibility to cripple the function
of the PRC2 complex by interfering with its multimeric integrity fueled
the interest in developing EZH2–EED protein–protein
interaction and EED inhibitors as indirect modulators of PRC2-dependent
methyltransferase activity. In this Perspective, we aim to summarize
the latest findings regarding the development and the biological activity
of these emerging classes of PRC2 modulators from a medicinal chemist’s
viewpoint.
Collapse
Affiliation(s)
- Stefano Tomassi
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy
| | - Annalisa Romanelli
- Department of Chemistry and Technology of Drugs, "Sapienza" University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Clemens Zwergel
- Department of Chemistry and Technology of Drugs, "Sapienza" University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Sergio Valente
- Department of Chemistry and Technology of Drugs, "Sapienza" University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Antonello Mai
- Department of Chemistry and Technology of Drugs, "Sapienza" University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| |
Collapse
|
11
|
Li X, Gera L, Zhang S, Chen Y, Lou L, Wilson LM, Xie ZR, Sautto G, Liu D, Danaher A, Mamouni K, Yang Y, Du Y, Fu H, Kucuk O, Osunkoya AO, Zhou J, Wu D. Pharmacological inhibition of noncanonical EED-EZH2 signaling overcomes chemoresistance in prostate cancer. Theranostics 2021; 11:6873-6890. [PMID: 34093859 PMCID: PMC8171087 DOI: 10.7150/thno.49235] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 04/22/2021] [Indexed: 12/25/2022] Open
Abstract
Rationale: Chemoresistance is a major obstacle in prostate cancer (PCa) treatment. We sought to understand the underlying mechanism of PCa chemoresistance and discover new treatments to overcome docetaxel resistance. Methods: We developed a novel phenotypic screening platform for the discovery of specific inhibitors of chemoresistant PCa cells. The mechanism of action of the lead compound was investigated using computational, molecular and cellular approaches. The in vivo toxicity and efficacy of the lead compound were evaluated in clinically-relevant animal models. Results: We identified LG1980 as a lead compound that demonstrates high selectivity and potency against chemoresistant PCa cells. Mechanistically, LG1980 binds embryonic ectoderm development (EED), disrupts the interaction between EED and enhancer of zeste homolog 2 (EZH2), thereby inducing the protein degradation of EZH2 and inhibiting the phosphorylation and activity of EZH2. Consequently, LG1980 targets a survival signaling cascade consisting of signal transducer and activator of transcription 3 (Stat3), S-phase kinase-associated protein 2 (SKP2), ATP binding cassette B 1 (ABCB1) and survivin. As a lead compound, LG1980 is well tolerated in mice and effectively suppresses the in vivo growth of chemoresistant PCa and synergistically enhances the efficacy of docetaxel in xenograft models. Conclusions: These results indicate that pharmacological inhibition of EED-EZH2 interaction is a novel strategy for the treatment of chemoresistant PCa. LG1980 and its analogues have the potential to be integrated into standard of care to improve clinical outcomes in PCa patients.
Collapse
Affiliation(s)
- Xin Li
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, USA
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Lajos Gera
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, School of Medicine, Aurora, CO, USA
| | - Shumin Zhang
- Department of Urology and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Yanhua Chen
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Lou
- School of Electrical and Computer Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - Lauren Marie Wilson
- School of Electrical and Computer Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - Zhong-Ru Xie
- School of Electrical and Computer Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - Giuseppe Sautto
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | | | - Alira Danaher
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, USA
| | - Kenza Mamouni
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Yang Yang
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuhong Du
- Department of Pharmacology and Chemical Biology, and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Haian Fu
- Department of Pharmacology and Chemical Biology, and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Omer Kucuk
- Department of Urology and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Adeboye O. Osunkoya
- Department of Urology and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
- Departments of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Daqing Wu
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, USA
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Urology and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
- MetCure Therapeutics LLC, Atlanta, GA, USA
| |
Collapse
|
12
|
Dockerill M, Gregson C, O' Donovan DH. Targeting PRC2 for the treatment of cancer: an updated patent review (2016 - 2020). Expert Opin Ther Pat 2021; 31:119-135. [PMID: 33103538 DOI: 10.1080/13543776.2021.1841167] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION PRC2 is a histone methyltransferase complex associated with several cancer types. Tazemetostat was recently approved as the first inhibitor targeting the catalytic subunit EZH2 and several other EZH2 inhibitors are now under clinical evaluation. Beyond EZH2, researchers have also explored other approaches including PRC2 activators, dual agents inhibiting both EZH1 and EZH2, allosteric inhibitors binding to EED, and compounds which induce the degradation of PRC2 constituent proteins. AREAS COVERED This review provides an overview of anticancer therapies targeting PRC2 during the period 2016-2020 including clinical trials, patents and the scientific literature. EXPERT OPINION The approval of tazemetostat marks the clinical validation of EZH2 for the treatment of cancer. Despite this success many questions remain; for instance, tazemetostat was briefly placed on clinical hold for safety concerns, while another EZH2 inhibitor (GSK126) demonstrated insufficient efficacy during a Phase I/II trial. It is important to understand these risks as PRC2 therapies progress through clinic evaluation. Alternative approaches to target PRC2 may offer distinct advantages over the inhibition of EZH2, including the potential to overcome EZH2 resistance mutations. However, these emerging modalities may also incur new challenges as they progress toward the clinic. Nonetheless, the diversity of agents under development represents a wealth of therapeutic options for future patients.
Collapse
|
13
|
Martin MC, Zeng G, Yu J, Schiltz GE. Small Molecule Approaches for Targeting the Polycomb Repressive Complex 2 (PRC2) in Cancer. J Med Chem 2020; 63:15344-15370. [PMID: 33283516 DOI: 10.1021/acs.jmedchem.0c01344] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The polycomb repressive complex 2 (PRC2) is composed of three core subunits, enhancer of zeste 2 (EZH2), embryonic ectoderm development (EED), and suppressor of zeste 12 (SUZ12), along with a number of accessory proteins. It is the key enzymatic protein complex that catalyzes histone H3 lysine 27 (H3K27) methylation to mediate epigenetic silencing of target genes. PRC2 thus plays essential roles in maintaining embryonic stem cell identity and in controlling cellular differentiation. Studies in the past decade have reported frequent overexpression or mutation of PRC2 in various cancers including prostate cancer and lymphoma. Aberrant PRC2 function has been extensively studied and proven to contribute to a large number of abnormal cellular processes, including those that lead to uncontrolled proliferation and tumorigenesis. Significant efforts have recently been made to develop small molecules targeting PRC2 function for potential use as anticancer therapeutics. In this review, we describe recent approaches to identify and develop small molecules that target PRC2. These various strategies include the inhibition of the function of individual PRC2 core proteins, the disruption of PRC2 complex formation, and the degradation of its subunits.
Collapse
Affiliation(s)
- M Cynthia Martin
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois 60208, United States
| | - Guihua Zeng
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Jindan Yu
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States.,Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Gary E Schiltz
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois 60208, United States.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States.,Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| |
Collapse
|
14
|
Rugo HS, Jacobs I, Sharma S, Scappaticci F, Paul TA, Jensen-Pergakes K, Malouf GG. The Promise for Histone Methyltransferase Inhibitors for Epigenetic Therapy in Clinical Oncology: A Narrative Review. Adv Ther 2020; 37:3059-3082. [PMID: 32445185 PMCID: PMC7467409 DOI: 10.1007/s12325-020-01379-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Indexed: 12/21/2022]
Abstract
Epigenetic processes are essential for normal development and the maintenance of tissue-specific gene expression in mammals. Changes in gene expression and malignant cellular transformation can result from disruption of epigenetic mechanisms, and global disruption in the epigenetic landscape is a key feature of cancer. The study of epigenetics in cancer has revealed that human cancer cells harbor both genetic alterations and epigenetic abnormalities that interplay at all stages of cancer development. Unlike genetic mutations, epigenetic aberrations are potentially reversible through epigenetic therapy, providing a therapeutically relevant treatment option. Histone methyltransferase inhibitors are emerging as an epigenetic therapy approach with great promise in the field of clinical oncology. The recent accelerated approval of the enhancer of zeste homolog 2 (EZH2; also known as histone-lysine N-methyltransferase EZH2) inhibitor tazemetostat for metastatic or locally advanced epithelioid sarcoma marks the first approval of such a compound for the treatment of cancer. Many other histone methyltransferase inhibitors are currently in development, some of which are being tested in clinical studies. This review focuses on histone methyltransferase inhibitors, highlighting their potential in the treatment of cancer. We also discuss the role for such epigenetic drugs in overcoming epigenetically driven drug resistance mechanisms, and their value in combination with other therapeutic approaches such as immunotherapy.
Collapse
|
15
|
Huang D, Tian S, Qi Y, Zhang JZH. Binding Modes of Small-Molecule Inhibitors to the EED Pocket of PRC2. Chemphyschem 2020; 21:263-271. [PMID: 31816138 DOI: 10.1002/cphc.201900903] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/30/2019] [Indexed: 12/30/2022]
Abstract
Polycomb Polycomb repressive complex 2 (PRC2) plays a key role in silencing epigenetic gene through trimethylation of lysine 27 on histone 3 (H3K27). Dysregulations of PRC2 caused by overexpression and mutations of the core subunits of PRC2 have been implicated in many cancers. The core subunits EZH1/2 are histone-lysine N-methyltransferases that function as the enzymatic component of PRC2. While the core subunit EED is a scaffolding protein to support EZH1/2 and binds JARID2K116me3/H3K27me3 to enhance the enzymatic activity of PRC2 through allosteric activation. Recently, several small molecules that compete with JARI2K116me3 and H3K27me3 have been reported. These molecules selectively bind to the JARID2K116me3/H3K27me3-binding pocket of EED, thereby preventing the allosteric regulation of PRC2. These first-in-class PRC2 inhibitors show robust suppression in DLBCL cell lines, demonstrating anticancer drugs that target the EED subunit of PRC2 are viable. In this study, we used the recently developed MM/GBSA_IE and the alanine scanning method to analyze the hot spots in EED/inhibitor interactions. The analysis of these hot and warm spots helps us to understand the fundamental differences between inhibitors. Our results give a quantitative explanation on why the binding affinities of EED/A-395 interactions are stronger than that of EED/EED226 while their binding modes are similar and provide valuable insights for rational design of novel EED inhibitors.
Collapse
Affiliation(s)
- Dading Huang
- School of Physics and Material Science, Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China
| | - Shuaizhen Tian
- School of Physics and Material Science, Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China
| | - Yifei Qi
- School of Physics and Material Science, Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China.,NYU-ECNU Center for Computational Chemistry at NYU Shanghai, Shanghai, 200062, China
| | - John Z H Zhang
- School of Physics and Material Science, Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China.,NYU-ECNU Center for Computational Chemistry at NYU Shanghai, Shanghai, 200062, China.,Department of Chemistry, New York University, NY, NY 10003, USA.,Collaborative Innovation Center of Extreme Optics, Shanxi University, Taiyuan, Shanxi, 030006, China
| |
Collapse
|
16
|
Li B, Chng WJ. EZH2 abnormalities in lymphoid malignancies: underlying mechanisms and therapeutic implications. J Hematol Oncol 2019; 12:118. [PMID: 31752930 PMCID: PMC6868783 DOI: 10.1186/s13045-019-0814-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 10/27/2019] [Indexed: 02/08/2023] Open
Abstract
EZH2 is the catalytic subunit of the polycomb repressive complex 2 (PRC2), which along with other PRC2 components mediates gene expression suppression via the methylation of Histone H3 at lysine 27. Recent studies have revealed a dichotomous role of EZH2 in physiology and in the pathogenesis of cancer. While it plays an essential role in the development of the lymphoid system, its deregulation, whether due to genetic or non-genetic causes, promotes B cell- and T cell-related lymphoma or leukemia. These findings triggered a boom in the development of therapeutic EZH2 inhibitors in recent years. Here, we discuss physiologic and pathogenic function of EZH2 in lymphoid context, various internal causes of EZH2 aberrance and how EZH2 modulates lymphomagenesis through epigenetic silencing, post-translational modifications (PTMs), orchestrating with surrounding tumor micro-environment and associating with RNA or viral partners. We also summarize different strategies to directly inhibit PRC2-EZH2 or to intervene EZH2 upstream signaling.
Collapse
Affiliation(s)
- Boheng Li
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Wee-Joo Chng
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore. .,Department of Haematology-Oncology, National University Cancer Institute of Singapore, Singapore, Singapore. .,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
17
|
Chammas P, Mocavini I, Di Croce L. Engaging chromatin: PRC2 structure meets function. Br J Cancer 2019; 122:315-328. [PMID: 31708574 PMCID: PMC7000746 DOI: 10.1038/s41416-019-0615-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/24/2019] [Indexed: 01/01/2023] Open
Abstract
Polycomb repressive complex 2 (PRC2) is a key epigenetic multiprotein complex involved in the regulation of gene expression in metazoans. PRC2 is formed by a tetrameric core that endows the complex with histone methyltransferase activity, allowing it to mono-, di- and tri-methylate histone H3 on lysine 27 (H3K27me1/2/3); H3K27me3 is a hallmark of facultative heterochromatin. The core complex of PRC2 is bound by several associated factors that are responsible for modulating its targeting specificity and enzymatic activity. Depletion and/or mutation of the subunits of this complex can result in severe developmental defects, or even lethality. Furthermore, mutations of these proteins in somatic cells can be drivers of tumorigenesis, by altering the transcriptional regulation of key tumour suppressors or oncogenes. In this review, we present the latest results from structural studies that have characterised PRC2 composition and function. We compare this information with data and literature for both gain-of function and loss-of-function missense mutations in cancers to provide an overview of the impact of these mutations on PRC2 activity.
Collapse
Affiliation(s)
- Paul Chammas
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Dr. Aiguader 88, Barcelona, 08003, Spain
| | - Ivano Mocavini
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Dr. Aiguader 88, Barcelona, 08003, Spain
| | - Luciano Di Croce
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Dr. Aiguader 88, Barcelona, 08003, Spain. .,Universitat Pompeu Fabra (UPF), Barcelona, Spain. .,ICREA, Pg Lluis Companys 23, Barcelona, 08010, Spain.
| |
Collapse
|
18
|
C10ORF12 modulates PRC2 histone methyltransferase activity and H3K27me3 levels. Acta Pharmacol Sin 2019; 40:1457-1465. [PMID: 31186533 DOI: 10.1038/s41401-019-0247-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/05/2019] [Indexed: 01/03/2023]
Abstract
The polycomb repressive complex 2 (PRC2) catalyzes the methylation of histone H3 on lysine 27 (H3K27) to generate trimethyl-H3K27 (H3K27me3) marks, thereby leading to a repressive chromatin state that inhibits gene expression. C10ORF12 was recently identified as a novel PRC2 interactor. Here, we show that C10ORF12 specifically interacts with PRC2 through its middle region (positions 619-718). C10ORF12 significantly enhances the histone methyltransferase activity of PRC2 in vitro and dramatically increases the total H3K27me3 levels in HeLa cells. C10ORF12 also antagonizes Jarid2, which is an auxiliary factor of the PRC2.2 subcomplex, to promote increased H3K27me3 levels in HeLa cells. Moreover, C10ORF12 alters the substrate preference of PRC2. These results indicate that C10ORF12 functions as a positive regulator of PRC2 and facilitates PRC2-mediated H3K27me3 modification of chromatin. These findings provide new insight into the roles of C10ORF12 in regulating the activity of the PRC2 complex.
Collapse
|
19
|
Read JA, Tart J, Rawlins PB, Gregson C, Jones K, Gao N, Zhu X, Tomlinson R, Code E, Cheung T, Chen H, Kawatkar SP, Bloecher A, Bagal S, O’Donovan DH, Robinson J. Rapid Identification of Novel Allosteric PRC2 Inhibitors. ACS Chem Biol 2019; 14:2134-2140. [PMID: 31525019 DOI: 10.1021/acschembio.9b00468] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Enhancer of zeste homologue 2 (EZH2), the catalytic subunit of polycomb repressive complex 2 (PRC2), regulates chromatin state and gene expression by methylating histone H3 lysine 27. EZH2 is overexpressed or mutated in various hematological malignancies and solid cancers. Our previous efforts to identify inhibitors of PRC2 methyltransferase activity by high-throughput screening (HTS) resulted in large numbers of false positives and thus a significant hit deconvolution challenge. More recently, others have reported compounds that bind to another PRC2 core subunit, EED, and allosterically inhibit EZH2 activity. This mechanism is particularly appealing as it appears to retain potency in cell lines that have acquired resistance to orthosteric EZH2 inhibition. By designing a fluorescence polarization probe based on the reported EED binding compounds, we were able to quickly and cleanly re-triage our previously challenging HTS hit list and identify novel allosteric PRC2 inhibitors.
Collapse
Affiliation(s)
- Jon A. Read
- Structure and Biophysics, Discovery Sciences, R&D, AstraZeneca, Cambridge, CB4 0WG U.K
| | - Jonathan Tart
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, CB4 0WG U.K
| | - Philip B. Rawlins
- Structure and Biophysics, Discovery Sciences, R&D, AstraZeneca, Cambridge, CB4 0WG U.K
| | - Clare Gregson
- Medicinal Chemistry, Oncology R&D, AstraZeneca, Cambridge, CB4 0WG U.K
| | - Karen Jones
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Macclesfield, SK10 4TG U.K
| | - Ning Gao
- Mechanistic Biology and Profiling, Discovery Sciences, R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Xiahui Zhu
- Mechanistic Biology and Profiling, Discovery Sciences, R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Ron Tomlinson
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Erin Code
- Mechanistic Biology and Profiling, Discovery Sciences, R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Tony Cheung
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Huawei Chen
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Sameer P. Kawatkar
- Medicinal Chemistry, Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Andy Bloecher
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Sharan Bagal
- Medicinal Chemistry, Oncology R&D, AstraZeneca, Cambridge, CB4 0WG U.K
| | | | - James Robinson
- Mechanistic Biology and Profiling, Discovery Sciences, R&D, AstraZeneca, Cambridge, CB4 0WG U.K
| |
Collapse
|
20
|
Kouznetsova VL, Tchekanov A, Li X, Yan X, Tsigelny IF. Polycomb repressive 2 complex-Molecular mechanisms of function. Protein Sci 2019; 28:1387-1399. [PMID: 31095801 PMCID: PMC6635771 DOI: 10.1002/pro.3647] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 05/13/2019] [Accepted: 05/14/2019] [Indexed: 12/31/2022]
Abstract
Numerous molecular processes conduct epigenetic regulation of protein transcription to maintain cell specification. In this review, we discuss molecular mechanisms of the Polycomb group of proteins and its enzymatic role in epigenetics. More specifically, we focus on the Polycomb repressive complex 2 (PRC2) and the effects of its repressive marker. We have compiled information regarding the biological structure and how that impacts the stability of the complex. In addition, we examined functions of the individual core proteins of PRC2 in relation to the accessory proteins that interact with the complex. Lastly, we discuss the implications of unregulated and downregulated PRC2 activity in Alzheimer's disease and cancer and possible methods of treatment related to PRC2 regulation.
Collapse
Affiliation(s)
- Valentina L. Kouznetsova
- Moores Cancer Center, UC San DiegoLa JollaCalifornia92093
- San Diego Supercomputer Center, UC San DiegoLa JollaCalifornia92093
| | | | - Xiaoming Li
- Saviour Bioscience, Inc.San DiegoCalifornia92121
| | | | - Igor F. Tsigelny
- Moores Cancer Center, UC San DiegoLa JollaCalifornia92093
- San Diego Supercomputer Center, UC San DiegoLa JollaCalifornia92093
- CureMatch, Inc.San DiegoCA 92121
| |
Collapse
|
21
|
Fioravanti R, Stazi G, Zwergel C, Valente S, Mai A. Six Years (2012-2018) of Researches on Catalytic EZH2 Inhibitors: The Boom of the 2-Pyridone Compounds. CHEM REC 2018; 18:1818-1832. [PMID: 30338896 PMCID: PMC7410397 DOI: 10.1002/tcr.201800091] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/26/2018] [Indexed: 12/20/2022]
Abstract
Enhancer of zeste homolog 2 (EZH2), the catalytic subunit of the Polycomb repressive complex 2 (PRC2), catalyzes the methylation of lysine 27 of histone H3 (H3K27) up to its trimethylated form (H3K27me), inducing by this way block of transcription and gene silencing. High levels of H3K27me3 have been found in both hematological malignancies and solid cancers, due to EZH2 overexpression and/or EZH2 mutation. From 2012, a number of highly potent and selective catalytic inhibitors of EZH2 have been reported, almost all bearing a 2-pyridone group in their structure. Typically, 2-pyridone inhibitors are selective for EZH2 over other methyltransferases, and some of them are specific for EZH2 over EZH1, others behave as dual EZH2/EZH1 inhibitors. The 2-pyridone moiety was crucial for the enzyme inhibition, as revealed later by crystallographic studies because it occupies partially the site for the co-substrate SAM (or the by-product, SAH) in the binding pocket of the enzyme, accounting for the SAM-competitive mechanism of action displayed by all the 2-pyridone inhibitors. The 2-pyridone warhead is linked to a support substructure, that can be either a bicyclic heteroaromatic ring (such as indazole, see for instance EPZ005687 and UNC1999, or indole, see for instance GSK126, EI1, and the more recent CPI-1205) or a simple monocyclic (hetero) aromatic ring (tazemetostat, MC3629, (R)-OR-S1/2), eventually annulated with the amide chain carrying the 2-pyridone group (3,4-dihydroisoquinoline-1(2H)-ones). Different substitutions at the support moiety influence the pharmacokinetics and pharmacodynamics of the compounds as well as their water solubility. In cancer diseases, the first reported 2-pyridone inhibitors displayed high antiproliferative effects in vitro and in vivo in lymphomas characterized by mutant EZH2 (such as Y641N), but the most recent compounds exert their anticancer activity against tumors with wild-type EZH2 as well. The dual EZH2/1 inhibitors have been recently reported to be more effective than EZH2 selective inhibitors in specific leukemias including leukemias cancer stem cells.
Collapse
Affiliation(s)
- Rossella Fioravanti
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le Aldo Moro n. 5., 00185, Roma, Italy
| | - Giulia Stazi
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le Aldo Moro n. 5., 00185, Roma, Italy
| | - Clemens Zwergel
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le Aldo Moro n. 5., 00185, Roma, Italy
| | - Sergio Valente
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le Aldo Moro n. 5., 00185, Roma, Italy
| | - Antonello Mai
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le Aldo Moro n. 5., 00185, Roma, Italy
| |
Collapse
|
22
|
Boriack-Sjodin PA, Ribich S, Copeland RA. RNA-modifying proteins as anticancer drug targets. Nat Rev Drug Discov 2018; 17:435-453. [PMID: 29773918 DOI: 10.1038/nrd.2018.71] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
All major biological macromolecules (DNA, RNA, proteins and lipids) undergo enzyme-catalysed covalent modifications that impact their structure, function and stability. A variety of covalent modifications of RNA have been identified and demonstrated to affect RNA stability and translation to proteins; these mechanisms of translational control have been termed epitranscriptomics. Emerging data suggest that some epitranscriptomic mechanisms are altered in human cancers as well as other human diseases. In this Review, we examine the current understanding of RNA modifications with a focus on mRNA methylation, highlight their possible roles in specific cancer indications and discuss the emerging potential of RNA-modifying proteins as therapeutic targets.
Collapse
|
23
|
Lee CH, Yu JR, Kumar S, Jin Y, LeRoy G, Bhanu N, Kaneko S, Garcia BA, Hamilton AD, Reinberg D. Allosteric Activation Dictates PRC2 Activity Independent of Its Recruitment to Chromatin. Mol Cell 2018; 70:422-434.e6. [PMID: 29681499 PMCID: PMC5935545 DOI: 10.1016/j.molcel.2018.03.020] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 02/20/2018] [Accepted: 03/16/2018] [Indexed: 12/12/2022]
Abstract
PRC2 is a therapeutic target for several types of cancers currently undergoing clinical trials. Its activity is regulated by a positive feedback loop whereby its terminal enzymatic product, H3K27me3, is specifically recognized and bound by an aromatic cage present in its EED subunit. The ensuing allosteric activation of the complex stimulates H3K27me3 deposition on chromatin. Here we report a stepwise feedback mechanism entailing key residues within distinctive interfacing motifs of EZH2 or EED that are found to be mutated in cancers and/or Weaver syndrome. PRC2 harboring these EZH2 or EED mutants manifested little activity in vivo but, unexpectedly, exhibited similar chromatin association as wild-type PRC2, indicating an uncoupling of PRC2 activity and recruitment. With genetic and chemical tools, we demonstrated that targeting allosteric activation overrode the gain-of-function effect of EZH2Y646X oncogenic mutations. These results revealed critical implications for the regulation and biology of PRC2 and a vulnerability in tackling PRC2-addicted cancers.
Collapse
Affiliation(s)
- Chul-Hwan Lee
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Jia-Ray Yu
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Sunil Kumar
- Department of Chemistry, New York University, New York, NY 10003, USA
| | - Ying Jin
- Shared Bioinformatics Core, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Gary LeRoy
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Natarajan Bhanu
- Department of Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Syuzo Kaneko
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Benjamin A Garcia
- Department of Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew D Hamilton
- Department of Chemistry, New York University, New York, NY 10003, USA
| | - Danny Reinberg
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
24
|
Senisterra G, Zhu HY, Luo X, Zhang H, Xun G, Lu C, Xiao W, Hajian T, Loppnau P, Chau I, Li F, Allali-Hassani A, Atadja P, Oyang C, Li E, Brown PJ, Arrowsmith CH, Zhao K, Yu Z, Vedadi M. Discovery of Small-Molecule Antagonists of the H3K9me3 Binding to UHRF1 Tandem Tudor Domain. SLAS DISCOVERY 2018; 23:930-940. [PMID: 29562800 DOI: 10.1177/2472555218766278] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ubiquitin-like with PHD and RING finger domains 1 (UHRF1) is a multidomain protein that plays a critical role in maintaining DNA methylation patterns through concurrent recognition of hemimethylated DNA and histone marks by various domains, and recruitment of DNA methyltransferase 1 (DNMT1). UHRF1 is overexpressed in various cancers, including breast cancer. The tandem tudor domain (TTD) of UHRF1 specifically and tightly binds to histone H3 di- or trimethylated at lysine 9 (H3K9me2 or H3K9me3, respectively), and this binding is essential for UHRF1 function. We developed an H3K9me3 peptide displacement assay, which was used to screen a library of 44,000 compounds for small molecules that disrupt the UHRF1-H3K9me3 interaction. This screen resulted in the identification of NV01, which bound to UHRF1-TTD with a Kd value of 5 μM. The structure of UHRF1-TTD in complex with NV01 confirmed binding to the H3K9me3-binding pocket. Limited structure-based optimization of NV01 led to the discovery of NV03 (Kd of 2.4 μM). These well-characterized small-molecule antagonists of the UHRF1-H3K9me2/3 interaction could be valuable starting chemical matter for developing more potent and cell-active probes toward further characterizing UHRF1 function, with possible applications as anticancer therapeutics.
Collapse
Affiliation(s)
| | - Hugh Y Zhu
- 2 Novartis Institutes for BioMedical Research (China), Pudong, Shanghai, China
| | - Xiao Luo
- 2 Novartis Institutes for BioMedical Research (China), Pudong, Shanghai, China
| | - Hailong Zhang
- 2 Novartis Institutes for BioMedical Research (China), Pudong, Shanghai, China
| | - Guoliang Xun
- 2 Novartis Institutes for BioMedical Research (China), Pudong, Shanghai, China
| | - Chunliang Lu
- 2 Novartis Institutes for BioMedical Research (China), Pudong, Shanghai, China
| | - Wen Xiao
- 2 Novartis Institutes for BioMedical Research (China), Pudong, Shanghai, China
| | - Taraneh Hajian
- 1 Structural Genomics Consortium, University of Toronto, Toronto, ON, Canada
| | - Peter Loppnau
- 1 Structural Genomics Consortium, University of Toronto, Toronto, ON, Canada
| | - Irene Chau
- 1 Structural Genomics Consortium, University of Toronto, Toronto, ON, Canada
| | - Fengling Li
- 1 Structural Genomics Consortium, University of Toronto, Toronto, ON, Canada
| | | | - Peter Atadja
- 2 Novartis Institutes for BioMedical Research (China), Pudong, Shanghai, China
| | - Counde Oyang
- 2 Novartis Institutes for BioMedical Research (China), Pudong, Shanghai, China
| | - En Li
- 2 Novartis Institutes for BioMedical Research (China), Pudong, Shanghai, China
| | - Peter J Brown
- 1 Structural Genomics Consortium, University of Toronto, Toronto, ON, Canada
| | - Cheryl H Arrowsmith
- 1 Structural Genomics Consortium, University of Toronto, Toronto, ON, Canada.,3 Princess Margaret Cancer Centre and Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Kehao Zhao
- 2 Novartis Institutes for BioMedical Research (China), Pudong, Shanghai, China.,4 School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Zhengtian Yu
- 2 Novartis Institutes for BioMedical Research (China), Pudong, Shanghai, China
| | - Masoud Vedadi
- 1 Structural Genomics Consortium, University of Toronto, Toronto, ON, Canada.,5 Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
25
|
Abstract
Dysregulation of the histone methyltransferase EZH2 plays a critical role in the development of a variety of malignancies including B-cell lymphomas. As a result, a series of small molecule inhibitors of EZH2 have been developed and studied in the pre-clinical setting. Three EZH2 inhibitors: tazemetostat (EPZ-6438), GSK2816126 and CPI-1205 have moved into phase I/phase II clinical trials in patients with non-Hodgkin lymphoma and genetically defined solid tumors. Early data from the tazemetostat trials indicate an acceptable safety profile and early signs of activity in diffuse large B-cell lymphoma and follicular lymphoma, including patients with EZH2 wild-type and mutant tumors. In this review, we present the rationale, key pre-clinical and early clinical findings of small molecule EZH2 inhibitors for use in lymphoma as well as future challenges and potential opportunities for combination therapies.
Collapse
Affiliation(s)
- Nitya Gulati
- a Division of Pediatric Hematology/Oncology, Department of Pediatrics , Weill Cornell Medical College , New York , NY , USA.,b Division of Pediatric Hematology/Oncology , Memorial Sloan Kettering Cancer Center , New York , NY , USA
| | - Wendy Béguelin
- c Division of Hematology/Oncology, Department of Medicine , Weill Cornell Medical College , New York , NY , USA
| | - Lisa Giulino-Roth
- a Division of Pediatric Hematology/Oncology, Department of Pediatrics , Weill Cornell Medical College , New York , NY , USA.,b Division of Pediatric Hematology/Oncology , Memorial Sloan Kettering Cancer Center , New York , NY , USA.,c Division of Hematology/Oncology, Department of Medicine , Weill Cornell Medical College , New York , NY , USA
| |
Collapse
|
26
|
Schapira M, Tyers M, Torrent M, Arrowsmith CH. WD40 repeat domain proteins: a novel target class? Nat Rev Drug Discov 2017; 16:773-786. [PMID: 29026209 PMCID: PMC5975957 DOI: 10.1038/nrd.2017.179] [Citation(s) in RCA: 212] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Antagonism of protein-protein interactions (PPIs) with small molecules is becoming more feasible as a therapeutic approach. Successful PPI inhibitors tend to target proteins containing deep peptide-binding grooves or pockets rather than the more common large, flat protein interaction surfaces. Here, we review one of the most abundant PPI domains in the human proteome, the WD40 repeat (WDR) domain, which has a central peptide-binding pocket and is a member of the β-propeller domain-containing protein family. Recently, two WDR domain-containing proteins, WDR5 and EED, as well as other β-propeller domains have been successfully targeted by potent, specific, cell-active, drug-like chemical probes. Could WDR domains be a novel target class for drug discovery? Although the research is at an early stage and therefore not clinically validated, cautious optimism is justified, as WDR domain-containing proteins are involved in multiple disease-associated pathways. The druggability and structural diversity of WDR domain binding pockets suggest that understanding how to target this prevalent domain class will open up areas of disease biology that have so far resisted drug discovery efforts.
Collapse
Affiliation(s)
- Matthieu Schapira
- Structural Genomics Consortium, University of Toronto, Toronto, ON M5G 1L7, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Mike Tyers
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3C 3J7, Canada
- Mount Sinai Hospital, The Lunenfeld-Tanenbaum Research Institute, Toronto, ON M5G 1X5, Canada
| | - Maricel Torrent
- Discovery Research, AbbVie, Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Cheryl H. Arrowsmith
- Structural Genomics Consortium, University of Toronto, Toronto, ON M5G 1L7, Canada
- Princess Margaret Cancer Centre and Department of Medical Biophysics, University of Toronto, 101 College St., Toronto, ON M5G 1L7, Canada
| |
Collapse
|
27
|
|
28
|
Baumgart SJ, Haendler B. Exploiting Epigenetic Alterations in Prostate Cancer. Int J Mol Sci 2017; 18:ijms18051017. [PMID: 28486411 PMCID: PMC5454930 DOI: 10.3390/ijms18051017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 05/04/2017] [Accepted: 05/04/2017] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer affects an increasing number of men worldwide and is a leading cause of cancer-associated deaths. Beside genetic mutations, many epigenetic alterations including DNA and histone modifications have been identified in clinical prostate tumor samples. They have been linked to aberrant activity of enzymes and reader proteins involved in these epigenetic processes, leading to the search for dedicated inhibitory compounds. In the wake of encouraging anti-tumor efficacy results in preclinical models, epigenetic modulators addressing different targets are now being tested in prostate cancer patients. In addition, the assessment of microRNAs as stratification biomarkers, and early clinical trials evaluating suppressor microRNAs as potential prostate cancer treatment are being discussed.
Collapse
Affiliation(s)
- Simon J Baumgart
- Drug Discovery, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany.
| | - Bernard Haendler
- Drug Discovery, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany.
| |
Collapse
|
29
|
Abstract
INTRODUCTION The histone methyltransferase EZH2 is the catalytic subunit of the PRC2 complex involved in H3K27 trimethylation. Aberrant PRC2 activity has been reported in several cancers and EZH2 overexpression has been associated with poor outcome in different tumors. EZH2 somatic mutations and deletions was found in lymphomas, myelodysplastic and myeloproliferative disorders and associated with higher H3K27me3 levels. Numerous chemical entities have been studied as EZH2 inhibitors in the recent years and some of them entered the cancer clinical arena. Areas covered: This review summarizes recent efforts in the drug development of EZH2 inhibitors reported in the patent literature covering the 2014-2016 period, and their potential use as therapeutics mainly in cancerous diseases. Expert opinion: Despite the number of compounds described, only a few of them entered the clinical arena. Moreover, most of the compounds developed share a common 2-pyridone ring pharmacophore. Recently, secondary mutants have been described to be resistant to the standard EZH2 inhibitors treatment. Based on these data a lot of effort is still required to find new chemical entities that inhibit EZH2 directly, or indirectly (via PRC2 disruption). Several issues are still to be settled, such as drug resistance and the importance of selectivity over EZH1 or somatic EZH2 mutants.
Collapse
Affiliation(s)
- Giulia Stazi
- a Dipartimento di Chimica e Tecnologie del Farmaco , Sapienza Università di Roma , Rome , Italy.,b Istituto Pasteur-Fondazione Cenci Bolognetti , Sapienza Università di Roma , Rome , Italy
| | - Clemens Zwergel
- a Dipartimento di Chimica e Tecnologie del Farmaco , Sapienza Università di Roma , Rome , Italy
| | - Antonello Mai
- a Dipartimento di Chimica e Tecnologie del Farmaco , Sapienza Università di Roma , Rome , Italy.,b Istituto Pasteur-Fondazione Cenci Bolognetti , Sapienza Università di Roma , Rome , Italy
| | - Sergio Valente
- a Dipartimento di Chimica e Tecnologie del Farmaco , Sapienza Università di Roma , Rome , Italy
| |
Collapse
|
30
|
Curtin ML, Pliushchev MA, Li HQ, Torrent M, Dietrich JD, Jakob CG, Zhu H, Zhao H, Wang Y, Ji Z, Clark RF, Sarris KA, Selvaraju S, Shaw B, Algire MA, He Y, Richardson PL, Sweis RF, Sun C, Chiang GG, Michaelides MR. SAR of amino pyrrolidines as potent and novel protein-protein interaction inhibitors of the PRC2 complex through EED binding. Bioorg Med Chem Lett 2017; 27:1576-1583. [DOI: 10.1016/j.bmcl.2017.02.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 02/10/2017] [Accepted: 02/14/2017] [Indexed: 12/22/2022]
|
31
|
Yang CY, Wang S. Allosteric Inactivation of Polycomb Repressive Complex 2 (PRC2) by Inhibiting Its Adapter Protein: Embryonic Ectodomain Development (EED). J Med Chem 2017; 60:2212-2214. [PMID: 28256832 DOI: 10.1021/acs.jmedchem.7b00287] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Aberrant PRC2 activity produces gene repressive epigenetic marks in multiple diseases and led to identification of Ezh2 as a drug target. Recent studies have shown that the epigenetic reader protein EED, associated with Ezh2 in PRC2, has an additional function to stimulate the PRC2 activity after binding to H3K27me3. Optimizing a compound known to block the H3K27me3 site in EED discovered by in-house screening, Novartis scientists have now produced a compound that shows durable tumor regression in a lymphoma xenograft model.
Collapse
Affiliation(s)
- Chao-Yie Yang
- Department of Internal Medicine, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Shaomeng Wang
- Department of Internal Medicine, University of Michigan , Ann Arbor, Michigan 48109, United States
| |
Collapse
|
32
|
Huang Y, Zhang J, Yu Z, Zhang H, Wang Y, Lingel A, Qi W, Gu J, Zhao K, Shultz MD, Wang L, Fu X, Sun Y, Zhang Q, Jiang X, Zhang J, Zhang C, Li L, Zeng J, Feng L, Zhang C, Liu Y, Zhang M, Zhang L, Zhao M, Gao Z, Liu X, Fang D, Guo H, Mi Y, Gabriel T, Dillon MP, Atadja P, Oyang C. Discovery of First-in-Class, Potent, and Orally Bioavailable Embryonic Ectoderm Development (EED) Inhibitor with Robust Anticancer Efficacy. J Med Chem 2017; 60:2215-2226. [DOI: 10.1021/acs.jmedchem.6b01576] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Ying Huang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Jeff Zhang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Zhengtian Yu
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Hailong Zhang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Youzhen Wang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Andreas Lingel
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Wei Qi
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Justin Gu
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Kehao Zhao
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Michael D. Shultz
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Long Wang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Xingnian Fu
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Yongfeng Sun
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Qiong Zhang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Xiangqing Jiang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Jiangwei Zhang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Chunye Zhang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Ling Li
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Jue Zeng
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Lijian Feng
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Chao Zhang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Yueqin Liu
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Man Zhang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Lijun Zhang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Mengxi Zhao
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Zhenting Gao
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Xianghui Liu
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Douglas Fang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Haibing Guo
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Yuan Mi
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Tobias Gabriel
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Michael P. Dillon
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Peter Atadja
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Counde Oyang
- Novartis Institutes for BioMedical Research, 4218 Jinke Road, Shanghai 201203, China
| |
Collapse
|
33
|
Li L, Feng L, Shi M, Zeng J, Chen Z, Zhong L, Huang L, Guo W, Huang Y, Qi W, Lu C, Li E, Zhao K, Gu J. Split luciferase-based biosensors for characterizing EED binders. Anal Biochem 2017; 522:37-45. [PMID: 28111304 DOI: 10.1016/j.ab.2017.01.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/13/2017] [Accepted: 01/18/2017] [Indexed: 12/22/2022]
Abstract
The EED (embryonic ectoderm development) subunit of the Polycomb repressive complex 2 (PRC2) plays an important role in the feed forward regulation of the PRC2 enzymatic activity. We recently identified a new class of allosteric PRC2 inhibitors that bind to the H3K27me3 pocket of EED. Multiple assays were developed and used to identify and characterize this type of PRC2 inhibitors. One of them is a genetically encoded EED biosensor based on the EED[G255D] mutant and the split firefly luciferase. This EED biosensor can detect the compound binding in the transfected cells and in the in vitro biochemical assays. Compared to other commonly used cellular assays, the EED biosensor assay has the advantage of shorter compound incubation with cells. The in vitro EED biosensor is much more sensitive than other label-free biophysical assays (e.g. DSF, ITC). Based on the crystal structure, the DSF data as well as the biosensor assay data, it's most likely that compound-induced increase in the luciferase activity of the EED[G255D] biosensor results from the decreased non-productive interactions between the EED subdomain and other subdomains within the biosensor construct. This new insight of the mechanism might help to broaden the use of the split luciferase based biosensors.
Collapse
Affiliation(s)
- Ling Li
- China Novartis Institutes for Biomedical Research, 4218 Jinke Road, Shanghai 201203, China.
| | - Lijian Feng
- China Novartis Institutes for Biomedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Minlong Shi
- China Novartis Institutes for Biomedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Jue Zeng
- China Novartis Institutes for Biomedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Zijun Chen
- China Novartis Institutes for Biomedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Li Zhong
- China Novartis Institutes for Biomedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Li Huang
- China Novartis Institutes for Biomedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Weihui Guo
- China Novartis Institutes for Biomedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Ying Huang
- China Novartis Institutes for Biomedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Wei Qi
- China Novartis Institutes for Biomedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Chris Lu
- China Novartis Institutes for Biomedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - En Li
- China Novartis Institutes for Biomedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Kehao Zhao
- China Novartis Institutes for Biomedical Research, 4218 Jinke Road, Shanghai 201203, China
| | - Justin Gu
- China Novartis Institutes for Biomedical Research, 4218 Jinke Road, Shanghai 201203, China.
| |
Collapse
|