101
|
da Motta LL, Ledaki I, Purshouse K, Haider S, De Bastiani MA, Baban D, Morotti M, Steers G, Wigfield S, Bridges E, Li JL, Knapp S, Ebner D, Klamt F, Harris AL, McIntyre A. The BET inhibitor JQ1 selectively impairs tumour response to hypoxia and downregulates CA9 and angiogenesis in triple negative breast cancer. Oncogene 2017; 36:122-132. [PMID: 27292261 PMCID: PMC5061082 DOI: 10.1038/onc.2016.184] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 03/22/2016] [Accepted: 04/15/2016] [Indexed: 12/16/2022]
Abstract
The availability of bromodomain and extra-terminal inhibitors (BETi) has enabled translational epigenetic studies in cancer. BET proteins regulate transcription by selectively recognizing acetylated lysine residues on chromatin. BETi compete with this process leading to both downregulation and upregulation of gene expression. Hypoxia enables progression of triple negative breast cancer (TNBC), the most aggressive form of breast cancer, partly by driving metabolic adaptation, angiogenesis and metastasis through upregulation of hypoxia-regulated genes (for example, carbonic anhydrase 9 (CA9) and vascular endothelial growth factor A (VEGF-A). Responses to hypoxia can be mediated epigenetically, thus we investigated whether BETi JQ1 could impair the TNBC response induced by hypoxia and exert anti-tumour effects. JQ1 significantly modulated 44% of hypoxia-induced genes, of which two-thirds were downregulated including CA9 and VEGF-A. JQ1 prevented HIF binding to the hypoxia response element in CA9 promoter, but did not alter HIF expression or activity, suggesting some HIF targets are BET-dependent. JQ1 reduced TNBC growth in vitro and in vivo and inhibited xenograft vascularization. These findings identify that BETi dually targets angiogenesis and the hypoxic response, an effective combination at reducing tumour growth in preclinical studies.
Collapse
Affiliation(s)
- L L da Motta
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Biochemistry/UFRGS, Porto Alegre, Brazil
- CAPES Foundation, Ministry of Education of Brazil, Brasilia, Brazil
| | - I Ledaki
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - K Purshouse
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - S Haider
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | | | - D Baban
- High Throughput Genomics, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - M Morotti
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - G Steers
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - S Wigfield
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - E Bridges
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - J-L Li
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Institute of Translational and Stratified Medicine, Plymouth University, Peninsula Schools of Medicine and Dentistry, Plymouth, UK
| | - S Knapp
- Nuffield Department of Clinical Medicine, Structural Genomics Consortium, University of Oxford, Oxford, UK
- Goethe University Frankfurt, Institute for Pharmaceutical Chemistry and Buchmann Institute for Life Sciences, Campus Riedberg, Frankfurt, Germany
| | - D Ebner
- Nuffield Department of Medicine, Target Discovery Institute (TDI), University of Oxford, Oxford, UK
| | - F Klamt
- Department of Biochemistry/UFRGS, Porto Alegre, Brazil
| | - A L Harris
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - A McIntyre
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Cancer Biology, Division of Cancer and Stem Cells, The University of Nottingham, Nottingham, UK
| |
Collapse
|
102
|
Cheng C, Diao H, Zhang F, Wang Y, Wang K, Wu R. Deciphering the mechanisms of selective inhibition for the tandem BD1/BD2 in the BET-bromodomain family. Phys Chem Chem Phys 2017; 19:23934-23941. [DOI: 10.1039/c7cp04608a] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The bromodomain and extra terminal domain (BET) family of bromodomains (BRDs) are well-known drug targets for many human diseases.
Collapse
Affiliation(s)
- Chunyan Cheng
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou 510006
- P. R. China
| | - Hongjuan Diao
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou 510006
- P. R. China
| | - Fan Zhang
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou 510006
- P. R. China
| | - Yongheng Wang
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou 510006
- P. R. China
| | - Kai Wang
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou 510006
- P. R. China
| | - Ruibo Wu
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou 510006
- P. R. China
| |
Collapse
|
103
|
Baud'huin M, Lamoureux F, Jacques C, Rodriguez Calleja L, Quillard T, Charrier C, Amiaud J, Berreur M, Brounais-LeRoyer B, Owen R, Reilly GC, Bradner JE, Heymann D, Ory B. Inhibition of BET proteins and epigenetic signaling as a potential treatment for osteoporosis. Bone 2017; 94:10-21. [PMID: 27669656 DOI: 10.1016/j.bone.2016.09.020] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 09/07/2016] [Accepted: 09/22/2016] [Indexed: 11/17/2022]
Abstract
Histone modifications are important for maintaining the transcription program. BET proteins, an important class of "histone reading proteins", have recently been described as essential in bone biology. This study presents the therapeutic opportunity of BET protein inhibition in osteoporosis. We find that the pharmacological BET protein inhibitor JQ1 rescues pathologic bone loss in a post-ovariectomy osteoporosis model by increasing the trabecular bone volume and restoring mechanical properties. The BET protein inhibition suppresses osteoclast differentiation and activity as well as the osteoblastogenesis in vitro. Moreover, we show that treated non-resorbing osteoclasts could still activate osteoblast differentiation. In addition, specific inhibition of BRD4 using RNA interference inhibits osteoclast differentiation but strongly activates osteoblast mineralization activity. Mechanistically, JQ1 inhibits expression of the master osteoclast transcription factor NFATc1 and the transcription factor of osteoblast Runx2. These findings strongly support that targeting epigenetic chromatin regulators such as BET proteins may offer a promising alternative for the treatment of bone-related disorders such as osteoporosis.
Collapse
Affiliation(s)
- Marc Baud'huin
- INSERM, UMR 957, équipe labellisée ligue 2012, 1 Rue Gaston Veil, 44035 Nantes, France; Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Université de Nantes, Nantes Atlantique Universités, EA3822, 1 Rue Gaston Veil, 44035 Nantes, France; Nantes University Hospital, Nantes, France
| | - François Lamoureux
- INSERM, UMR 957, équipe labellisée ligue 2012, 1 Rue Gaston Veil, 44035 Nantes, France; Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Université de Nantes, Nantes Atlantique Universités, EA3822, 1 Rue Gaston Veil, 44035 Nantes, France
| | - Camille Jacques
- INSERM, UMR 957, équipe labellisée ligue 2012, 1 Rue Gaston Veil, 44035 Nantes, France; Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Université de Nantes, Nantes Atlantique Universités, EA3822, 1 Rue Gaston Veil, 44035 Nantes, France
| | - Lidia Rodriguez Calleja
- INSERM, UMR 957, équipe labellisée ligue 2012, 1 Rue Gaston Veil, 44035 Nantes, France; Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Université de Nantes, Nantes Atlantique Universités, EA3822, 1 Rue Gaston Veil, 44035 Nantes, France
| | - Thibaut Quillard
- INSERM, UMR 957, équipe labellisée ligue 2012, 1 Rue Gaston Veil, 44035 Nantes, France; Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Université de Nantes, Nantes Atlantique Universités, EA3822, 1 Rue Gaston Veil, 44035 Nantes, France
| | - Céline Charrier
- INSERM, UMR 957, équipe labellisée ligue 2012, 1 Rue Gaston Veil, 44035 Nantes, France; Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Université de Nantes, Nantes Atlantique Universités, EA3822, 1 Rue Gaston Veil, 44035 Nantes, France
| | - Jérome Amiaud
- INSERM, UMR 957, équipe labellisée ligue 2012, 1 Rue Gaston Veil, 44035 Nantes, France; Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Université de Nantes, Nantes Atlantique Universités, EA3822, 1 Rue Gaston Veil, 44035 Nantes, France
| | - Martine Berreur
- INSERM, UMR 957, équipe labellisée ligue 2012, 1 Rue Gaston Veil, 44035 Nantes, France; Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Université de Nantes, Nantes Atlantique Universités, EA3822, 1 Rue Gaston Veil, 44035 Nantes, France
| | - Bénédicte Brounais-LeRoyer
- INSERM, UMR 957, équipe labellisée ligue 2012, 1 Rue Gaston Veil, 44035 Nantes, France; Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Université de Nantes, Nantes Atlantique Universités, EA3822, 1 Rue Gaston Veil, 44035 Nantes, France
| | - Robert Owen
- Department of Materials Science and Engineering, INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
| | - Gwendolen C Reilly
- Department of Materials Science and Engineering, INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
| | - James E Bradner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA; Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Dominique Heymann
- INSERM, UMR 957, équipe labellisée ligue 2012, 1 Rue Gaston Veil, 44035 Nantes, France; Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Université de Nantes, Nantes Atlantique Universités, EA3822, 1 Rue Gaston Veil, 44035 Nantes, France; Nantes University Hospital, Nantes, France
| | - Benjamin Ory
- INSERM, UMR 957, équipe labellisée ligue 2012, 1 Rue Gaston Veil, 44035 Nantes, France; Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Université de Nantes, Nantes Atlantique Universités, EA3822, 1 Rue Gaston Veil, 44035 Nantes, France.
| |
Collapse
|
104
|
Nicholas DA, Andrieu G, Strissel KJ, Nikolajczyk BS, Denis GV. BET bromodomain proteins and epigenetic regulation of inflammation: implications for type 2 diabetes and breast cancer. Cell Mol Life Sci 2017; 74:231-243. [PMID: 27491296 PMCID: PMC5222701 DOI: 10.1007/s00018-016-2320-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/16/2016] [Accepted: 07/29/2016] [Indexed: 12/18/2022]
Abstract
Chronic inflammation drives pathologies associated with type 2 diabetes (T2D) and breast cancer. Obesity-driven inflammation may explain increased risk and mortality of breast cancer with T2D reported in the epidemiology literature. Therapeutic approaches to target inflammation in both T2D and cancer have so far fallen short of the expected improvements in disease pathogenesis or outcomes. The targeting of epigenetic regulators of cytokine transcription and cytokine signaling offers one promising, untapped approach to treating diseases driven by inflammation. Recent work has deeply implicated the Bromodomain and Extra-Terminal domain (BET) proteins, which are acetylated histone "readers", in epigenetic regulation of inflammation. This review focuses on inflammation associated with T2D and breast cancer, and the possibility of targeting BET proteins as an approach to regulating inflammation in the clinic. Understanding inflammation in the context of BET protein regulation may provide a basis for designing promising therapeutics for T2D and breast cancer.
Collapse
Affiliation(s)
- Dequina A Nicholas
- Cancer Center, Boston University School of Medicine, 72 East Concord Street, Room K520, Boston, MA, 02118, USA
- Department of Microbiology, Training Program in Inflammatory Disorders, 72 East Concord Street, K520, Boston, MA, 02118, USA
| | - Guillaume Andrieu
- Cancer Center, Boston University School of Medicine, 72 East Concord Street, Room K520, Boston, MA, 02118, USA
| | - Katherine J Strissel
- Cancer Center, Boston University School of Medicine, 72 East Concord Street, Room K520, Boston, MA, 02118, USA
| | - Barbara S Nikolajczyk
- Department of Microbiology, Training Program in Inflammatory Disorders, 72 East Concord Street, K520, Boston, MA, 02118, USA
| | - Gerald V Denis
- Cancer Center, Boston University School of Medicine, 72 East Concord Street, Room K520, Boston, MA, 02118, USA.
- Section of Hematology/Oncology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, 72 East Concord Street, K520, Boston, MA, 02118, USA.
| |
Collapse
|
105
|
Zhou B, Mu J, Gong Y, Lu C, Zhao Y, He T, Qin Z. Brd4 inhibition attenuates unilateral ureteral obstruction-induced fibrosis by blocking TGF-β-mediated Nox4 expression. Redox Biol 2016; 11:390-402. [PMID: 28063381 PMCID: PMC5219604 DOI: 10.1016/j.redox.2016.12.031] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 12/20/2016] [Accepted: 12/29/2016] [Indexed: 01/09/2023] Open
Abstract
Uncovering new therapeutic targets for renal fibrosis holds promise for the treatment of chronic kidney diseases. Bromodomain and extra-terminal (BET) protein inhibitors have been shown to effectively ameliorate pathological fibrotic responses. However, the pharmacological effects and underlying mechanisms of these inhibitors in renal fibrosis remain elusive. In this study, we determined that the inhibition of Brd4, a BET family member, with a selective potent chemical inhibitor, JQ1, could prevent the development of renal fibrosis and block the progression of fibrosis in rats that have undergone unilateral ureteral obstruction (UUO). Inhibiting Brd4 with either JQ1 or genetic knockdown resulted in decreased expression of fibrotic genes such as α-smooth muscle actin, collagen IV and fibronectin both in UUO-induced fibrosis and upon TGF-β1 stimulation in HK-2 cells. Brd4 inhibition also suppressed the oxidative stress induced by UUO in vivo or by TGF-β1 in HK-2 cells. Moreover, Nox4, which is constitutively active in renal cells and is involved in the generation of hydrogen peroxide, was up-regulated during UUO-mediated fibrosis and induced by TGF-β1 in HK-2 cells, and this up-regulation could be blunted by Brd4 inhibition. Consistently, Nox4-mediated ROS generation and fibrotic gene expression were attenuated upon Brd4 inhibition. Further, the transcriptional activity of Nox4 was suppressed by JQ1 or siRNA against Brd4. Additionally, Smad3 and ERK1/2 phosphorylation, which are upstream signals of Nox4 expression, were inhibited both in JQ1-administered UUO rats and Brd4-inhibited HK-2 cells. In conclusion, these results indicated that the inhibition of Brd4 might protect against renal fibrosis by blocking the TGF-β-Nox4-ROS-fibrosis axis, suggesting that Brd4 could be a promising therapeutic target. Brd4 was up-regulated in the progression of renal fibrosis. Brd4 inhibitor JQ1 prevented renal fibrosis and delayed the fibrotic progression. Brd4 inhibition blocked TGF-β1-induced oxidative stress and fibrosis through Nox4. Brd4 regulated Nox4 expression via Smad and ERK pathways.
Collapse
Affiliation(s)
- Baoshang Zhou
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Jiao Mu
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Yi Gong
- Department of Nephrology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Caibao Lu
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Youguang Zhao
- Department of Urology, Chengdu Military General Hospital, Chengdu 610083, China
| | - Ting He
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Zhexue Qin
- Department of Cardiology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| |
Collapse
|
106
|
Camus V, Tilly H. Managing early failures with R-CHOP in patients with diffuse large B-cell lymphoma. Expert Rev Hematol 2016; 10:1047-1055. [DOI: 10.1080/17474086.2016.1254547] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Vincent Camus
- Department of Hematology, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France
| | - Hervé Tilly
- Department of Hematology, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France
| |
Collapse
|
107
|
Xiong C, Masucci MV, Zhou X, Liu N, Zang X, Tolbert E, Zhao TC, Zhuang S. Pharmacological targeting of BET proteins inhibits renal fibroblast activation and alleviates renal fibrosis. Oncotarget 2016; 7:69291-69308. [PMID: 27732564 PMCID: PMC5342478 DOI: 10.18632/oncotarget.12498] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 09/29/2016] [Indexed: 12/27/2022] Open
Abstract
Bromodomain and extra-terminal (BET) protein inhibitors have been shown to effectively inhibit tumorgenesis and ameliorate pulmonary fibrosis by targeting bromodomain proteins that bind acetylated chromatin markers. However, their pharmacological effects in renal fibrosis remain unclear. In this study, we examined the effect of I-BET151, a selective and potent BET inhibitor, on renal fibroblast activation and renal fibrosis. In cultured renal interstitial fibroblasts, exposure of cells to I-BET151, or silencing of bromodoma in-containing protein 4 (Brd4), a key BET protein isoform, significantly reduced their activation as indicated by decreased expression of α-smooth muscle actin, collagen 1 and fibronectin. In a murine model of renal fibrosis induced by unilateral ureteral obstruction (UUO), administration of I-BET151 suppressed the deposition of extracellular matrix proteins, renal fibroblast activation and macrophage infiltration. Mechanistically, I-BET151 treatment abrogated UUO-induced phosphorylation of epidermal growth factor receptor and platelet growth factor receptor-β. It also inhibited the activation of Smad-3, STAT3 and NF-κB pathways, as well as the expression of c-Myc and P53 transcription factors in the kidney. Moreover, BET inhibition resulted in the reduction of renal epithelial cells arrested at the G2/M phase of cell cycle after UUO injury. Finally, injury to the kidney up-regulated Brd4, and I-BET151 treatment abrogated its expression. Brd4 was also highly expressed in human fibrotic kidneys. These data indicate that BET proteins are implicated in the regulation of signaling pathways and transcription factors associated with renal fibrogenesis, and suggest that pharmacological inhibition of BET proteins could be a potential treatment for renal fibrosis.
Collapse
Affiliation(s)
- Chongxiang Xiong
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, USA
| | - Monica V. Masucci
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, USA
| | - Xiaoxu Zhou
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, USA
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiujuan Zang
- Department of Nephrology, Shanghai Songjiang District Central Hospital, Shanghai, China
| | - Evelyn Tolbert
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, USA
| | - Ting C. Zhao
- Department of Surgery, Roger Williams Medical Center, Boston University, Providence, RI, USA
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, USA
| |
Collapse
|
108
|
Dickson BM, de Waal PW, Ramjan ZH, Xu HE, Rothbart SB. A fast, open source implementation of adaptive biasing potentials uncovers a ligand design strategy for the chromatin regulator BRD4. J Chem Phys 2016; 145:154113. [PMID: 27782467 PMCID: PMC5074994 DOI: 10.1063/1.4964776] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/30/2016] [Indexed: 12/17/2022] Open
Abstract
In this communication we introduce an efficient implementation of adaptive biasing that greatly improves the speed of free energy computation in molecular dynamics simulations. We investigated the use of accelerated simulations to inform on compound design using a recently reported and clinically relevant inhibitor of the chromatin regulator BRD4 (bromodomain-containing protein 4). Benchmarking on our local compute cluster, our implementation achieves up to 2.5 times more force calls per day than plumed2. Results of five 1 μs-long simulations are presented, which reveal a conformational switch in the BRD4 inhibitor between a binding competent and incompetent state. Stabilization of the switch led to a -3 kcal/mol improvement of absolute binding free energy. These studies suggest an unexplored ligand design principle and offer new actionable hypotheses for medicinal chemistry efforts against this druggable epigenetic target class.
Collapse
Affiliation(s)
- Bradley M Dickson
- Center for Epigenetics, Van Andel Research Institute, 333 Bostwick Avenue, NE, Grand Rapids, Michigan 49503, USA
| | - Parker W de Waal
- Laboratory of Structural Sciences, Van Andel Research Institute, 333 Bostwick Avenue, NE, Grand Rapids, Michigan 49503, USA
| | - Zachary H Ramjan
- Center for Epigenetics, Van Andel Research Institute, 333 Bostwick Avenue, NE, Grand Rapids, Michigan 49503, USA
| | - H Eric Xu
- Laboratory of Structural Sciences, Van Andel Research Institute, 333 Bostwick Avenue, NE, Grand Rapids, Michigan 49503, USA
| | - Scott B Rothbart
- Center for Epigenetics, Van Andel Research Institute, 333 Bostwick Avenue, NE, Grand Rapids, Michigan 49503, USA
| |
Collapse
|
109
|
Maiuri AR, O'Hagan HM. Interplay Between Inflammation and Epigenetic Changes in Cancer. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 144:69-117. [PMID: 27865469 DOI: 10.1016/bs.pmbts.2016.09.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Immune responses can suppress tumorigenesis, but also contribute to cancer initiation and progression suggesting a complex interaction between the immune system and cancer. Epigenetic alterations, which are heritable changes in gene expression without changes to the DNA sequence, also play a role in carcinogenesis through silencing expression of tumor suppressor genes and activating oncogenic signaling. Interestingly, epithelial cells at sites of chronic inflammation undergo DNA methylation alterations that are similar to those present in cancer cells, suggesting that inflammation may initiate cancer-specific epigenetic changes in epithelial cells. Furthermore, epigenetic changes occur during immune cell differentiation and participate in regulating the immune response, including the regulation of inflammatory cytokines. Cancer cells utilize epigenetic silencing of immune-related genes to evade the immune response. This chapter will detail the interactions between inflammation and epigenetics in tumor initiation, promotion, and immune evasion and how these connections are being leveraged in cancer prevention and treatment.
Collapse
Affiliation(s)
- A R Maiuri
- Medical Sciences, Indiana University School of Medicine, Bloomington, IN, United States
| | - H M O'Hagan
- Medical Sciences, Indiana University School of Medicine, Bloomington, IN, United States; Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, United States.
| |
Collapse
|
110
|
Meslamani J, Smith SG, Sanchez R, Zhou MM. Structural features and inhibitors of bromodomains. DRUG DISCOVERY TODAY. TECHNOLOGIES 2016; 19:3-15. [PMID: 27769355 DOI: 10.1016/j.ddtec.2016.09.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 07/12/2016] [Accepted: 09/06/2016] [Indexed: 01/02/2023]
Abstract
Bromodomains are conserved structural modules responsible for recognizing acetylated-lysine residues on histone tails and other transcription-associated proteins, such as transcription factors and co-factors. Owing to their important functions in the regulation of ordered gene transcription in chromatin, bromodomains of the BET family proteins have recently been shown as druggable targets for a wide array of human diseases, including cancer and inflammation. Here we review the structural and functional features of the bromodomains and their small-molecule inhibitors. Additional new insights provided herein highlight the landscape of the ligand binding sites in the bromodomains that will hopefully facilitate further development of new inhibitors with optimal affinity and selectivity.
Collapse
Affiliation(s)
- Jamel Meslamani
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, United States.
| | - Steven G Smith
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, United States
| | - Roberto Sanchez
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, United States
| | - Ming-Ming Zhou
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, United States
| |
Collapse
|
111
|
Gong F, Chiu LY, Miller KM. Acetylation Reader Proteins: Linking Acetylation Signaling to Genome Maintenance and Cancer. PLoS Genet 2016; 12:e1006272. [PMID: 27631103 PMCID: PMC5025232 DOI: 10.1371/journal.pgen.1006272] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chromatin-based DNA damage response (DDR) pathways are fundamental for preventing genome and epigenome instability, which are prevalent in cancer. Histone acetyltransferases (HATs) and histone deacetylases (HDACs) catalyze the addition and removal of acetyl groups on lysine residues, a post-translational modification important for the DDR. Acetylation can alter chromatin structure as well as function by providing binding signals for reader proteins containing acetyl-lysine recognition domains, including the bromodomain (BRD). Acetylation dynamics occur upon DNA damage in part to regulate chromatin and BRD protein interactions that mediate key DDR activities. In cancer, DDR and acetylation pathways are often mutated or abnormally expressed. DNA damaging agents and drugs targeting epigenetic regulators, including HATs, HDACs, and BRD proteins, are used or are being developed to treat cancer. Here, we discuss how histone acetylation pathways, with a focus on acetylation reader proteins, promote genome stability and the DDR. We analyze how acetylation signaling impacts the DDR in the context of cancer and its treatments. Understanding the relationship between epigenetic regulators, the DDR, and chromatin is integral for obtaining a mechanistic understanding of genome and epigenome maintenance pathways, information that can be leveraged for targeting acetylation signaling, and/or the DDR to treat diseases, including cancer.
Collapse
Affiliation(s)
- Fade Gong
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, United States of America
| | - Li-Ya Chiu
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, United States of America
| | - Kyle M. Miller
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, United States of America
- * E-mail:
| |
Collapse
|
112
|
Uitdehaag JCM, de Roos JADM, Prinsen MBW, Willemsen-Seegers N, de Vetter JRF, Dylus J, van Doornmalen AM, Kooijman J, Sawa M, van Gerwen SJC, de Man J, Buijsman RC, Zaman GJR. Cell Panel Profiling Reveals Conserved Therapeutic Clusters and Differentiates the Mechanism of Action of Different PI3K/mTOR, Aurora Kinase and EZH2 Inhibitors. Mol Cancer Ther 2016; 15:3097-3109. [PMID: 27587489 DOI: 10.1158/1535-7163.mct-16-0403] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/08/2016] [Accepted: 08/21/2016] [Indexed: 11/16/2022]
Abstract
Cancer cell line panels are important tools to characterize the in vitro activity of new investigational drugs. Here, we present the inhibition profiles of 122 anticancer agents in proliferation assays with 44 or 66 genetically characterized cancer cell lines from diverse tumor tissues (Oncolines). The library includes 29 cytotoxics, 68 kinase inhibitors, and 11 epigenetic modulators. For 38 compounds this is the first comparative profiling in a cell line panel. By strictly maintaining optimized assay protocols, biological variation was kept to a minimum. Replicate profiles of 16 agents over three years show a high average Pearson correlation of 0.8 using IC50 values and 0.9 using GI50 values. Good correlations were observed with other panels. Curve fitting appears a large source of variation. Hierarchical clustering revealed 44 basic clusters, of which 26 contain compounds with common mechanisms of action, of which 9 were not reported before, including TTK, BET and two clusters of EZH2 inhibitors. To investigate unexpected clusterings, sets of BTK, Aurora and PI3K inhibitors were profiled in biochemical enzyme activity assays and surface plasmon resonance binding assays. The BTK inhibitor ibrutinib clusters with EGFR inhibitors, because it cross-reacts with EGFR. Aurora kinase inhibitors separate into two clusters, related to Aurora A or pan-Aurora selectivity. Similarly, 12 inhibitors in the PI3K/AKT/mTOR pathway separated into different clusters, reflecting biochemical selectivity (pan-PI3K, PI3Kβγδ-isoform selective or mTOR-selective). Of these, only allosteric mTOR inhibitors preferentially targeted PTEN-mutated cell lines. This shows that cell line profiling is an excellent tool for the unbiased classification of antiproliferative compounds. Mol Cancer Ther; 15(12); 3097-109. ©2016 AACR.
Collapse
Affiliation(s)
- Joost C M Uitdehaag
- Netherlands Translational Research Center B.V., Kloosterstraat, the Netherlands
| | | | - Martine B W Prinsen
- Netherlands Translational Research Center B.V., Kloosterstraat, the Netherlands
| | | | | | - Jelle Dylus
- Netherlands Translational Research Center B.V., Kloosterstraat, the Netherlands
| | | | - Jeffrey Kooijman
- Netherlands Translational Research Center B.V., Kloosterstraat, the Netherlands
| | | | | | - Jos de Man
- Netherlands Translational Research Center B.V., Kloosterstraat, the Netherlands
| | - Rogier C Buijsman
- Netherlands Translational Research Center B.V., Kloosterstraat, the Netherlands
| | - Guido J R Zaman
- Netherlands Translational Research Center B.V., Kloosterstraat, the Netherlands.
| |
Collapse
|
113
|
Zeidan AM, Stahl M, Komrokji R. Emerging biological therapies for the treatment of myelodysplastic syndromes. Expert Opin Emerg Drugs 2016; 21:283-300. [DOI: 10.1080/14728214.2016.1220534] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
114
|
Raj U, Kumar H, Varadwaj PK. Molecular docking and dynamics simulation study of flavonoids as BET bromodomain inhibitors. J Biomol Struct Dyn 2016; 35:2351-2362. [PMID: 27494802 DOI: 10.1080/07391102.2016.1217276] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Bromodomains (BRDs) are the epigenetic proteins responsible for transcriptional regulation through its interaction with methylated or acetylated histone residues. The lysine residues of Bromodomain-1 (BD1) of Brd4 undergo ε-N-Acetylation posttranslational modifications to control transcription of genes. Due to its role in diverse cellular functions, Brd4 of bromodomain family, was considered as a prominent target for many diseases such as cancer, obesity, kidney disease, lung fibrosis, inflammatory diseases, etc. In this study, an attempt has been made to screen compounds from flavonoids and extended flavonoids libraries targeting acetylated lysine (KAc) binding site of BD1 of Brd4 using docking and molecular dynamics simulations. Two different docking programs AutoDock and Glide were used to compare their suitability for the receptor. Interestingly, in both the docking programs, the screened flavonoids have occupied the same binding pocket confirming the selection of active site. Further the MMGBSA binding free energy calculations and ADME analysis were carried out on screened compounds to establish their anti-cancerous properties. We have identified a flavonoid which shows docking and Glide e-model score comparatively much higher than those of already reported known inhibitors against Brd4. The protein-ligand complex with top-ranked flavonoid was used for dynamics simulation study for 50 ns in order to validate its stability inside the active site of Brd4 receptor. The results provide valuable information for structure-based drug design of Brd4 inhibitors.
Collapse
Affiliation(s)
- Utkarsh Raj
- a Department of Bioinformatics , Indian Institute of Information Technology-Allahabad , CC2-4203, Jhalwa Campus, Deoghat, Allahabad - 211012 , Uttar Pradesh , India
| | - Himansu Kumar
- a Department of Bioinformatics , Indian Institute of Information Technology-Allahabad , CC2-4203, Jhalwa Campus, Deoghat, Allahabad - 211012 , Uttar Pradesh , India
| | - Pritish Kumar Varadwaj
- a Department of Bioinformatics , Indian Institute of Information Technology-Allahabad , CC2-4203, Jhalwa Campus, Deoghat, Allahabad - 211012 , Uttar Pradesh , India
| |
Collapse
|
115
|
Suarez-Alvarez B, Morgado-Pascual JL, Rayego-Mateos S, Rodriguez RM, Rodrigues-Diez R, Cannata-Ortiz P, Sanz AB, Egido J, Tharaux PL, Ortiz A, Lopez-Larrea C, Ruiz-Ortega M. Inhibition of Bromodomain and Extraterminal Domain Family Proteins Ameliorates Experimental Renal Damage. J Am Soc Nephrol 2016; 28:504-519. [PMID: 27436852 DOI: 10.1681/asn.2015080910] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 06/05/2016] [Indexed: 12/31/2022] Open
Abstract
Renal inflammation has a key role in the onset and progression of immune- and nonimmune-mediated renal diseases. Therefore, the search for novel anti-inflammatory pharmacologic targets is of great interest in renal pathology. JQ1, a small molecule inhibitor of bromodomain and extraterminal (BET) proteins, was previously found to preserve renal function in experimental polycystic kidney disease. We report here that JQ1-induced BET inhibition modulated the in vitro expression of genes involved in several biologic processes, including inflammation and immune responses. Gene silencing of BRD4, an important BET protein, and chromatin immunoprecipitation assays showed that JQ1 alters the direct association of BRD4 with acetylated histone-packaged promoters and reduces the transcription of proinflammatory genes (IL-6, CCL-2, and CCL-5). In vivo, JQ1 abrogated experimental renal inflammation in murine models of unilateral ureteral obstruction, antimembrane basal GN, and infusion of Angiotensin II. Notably, JQ1 downregulated the expression of several genes controlled by the NF-κB pathway, a key inflammatory signaling pathway. The RelA NF-κB subunit is activated by acetylation of lysine 310. In damaged kidneys and cytokine-stimulated renal cells, JQ1 reduced the nuclear levels of RelA NF-κB. Additionally, JQ1 dampened the activation of the Th17 immune response in experimental renal damage. Our results show that inhibition of BET proteins reduces renal inflammation by several mechanisms: chromatin remodeling in promoter regions of specific genes, blockade of NF-κB pathway activation, and modulation of the Th17 immune response. These results suggest that inhibitors of BET proteins could have important therapeutic applications in inflammatory renal diseases.
Collapse
Affiliation(s)
| | | | | | - Ramon M Rodriguez
- Immunology Department, Hospital Universitario Central de Asturias, REDINREN, Oviedo, Spain
| | - Raul Rodrigues-Diez
- Nephrology Department, Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, REDINREN, Madrid, Spain
| | | | - Ana B Sanz
- Dialysis Unit, Instituto de Investigación Sanitaria (IIS) Fundación Jiménez Díaz, Nephrology Department, School of Medicine, Universidad Autónoma Madrid, Renal Research Retics (REDINREN), Madrid, Spain
| | - Jesus Egido
- IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma Madrid, Madrid, Spain; and
| | - Pierre-Louis Tharaux
- Paris Cardiovascular Research Centre (PARCC), Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Alberto Ortiz
- Dialysis Unit, Instituto de Investigación Sanitaria (IIS) Fundación Jiménez Díaz, Nephrology Department, School of Medicine, Universidad Autónoma Madrid, Renal Research Retics (REDINREN), Madrid, Spain
| | - Carlos Lopez-Larrea
- Immunology Department, Hospital Universitario Central de Asturias, REDINREN, Oviedo, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, Nephrology Department and
| |
Collapse
|
116
|
Scarfò I, Pellegrino E, Mereu E, Inghirami G, Piva R. Transposable elements: The enemies within. Exp Hematol 2016; 44:913-6. [PMID: 27377925 DOI: 10.1016/j.exphem.2016.06.251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 06/21/2016] [Accepted: 06/22/2016] [Indexed: 12/28/2022]
Abstract
Understanding transformation mechanisms other than genetic aberrations has recently captured the attention of cancer researchers. To date, the role of transposable elements (TEs) in tumor development remains largely undefined. However, an increasing number of studies have reported that loss of epigenetic control causes TE reactivation and consequent oncogenic transcription. Here, we discuss principal examples of TEs-driven oncogenesis. Available data suggest that long terminal repeats and long interspersed nuclear elements play a pivotal role as alternative promoters. These findings provide definitive experimental evidence that repetitive elements are a powerful underestimated force toward oncogenesis and open the possibility to new therapeutic treatments.
Collapse
Affiliation(s)
- Irene Scarfò
- Department of Molecular Biotechnology and Health Sciences; Center for Experimental Research and Medical Studies, University of Torino, Torino, Italy
| | - Elisa Pellegrino
- Department of Molecular Biotechnology and Health Sciences; Center for Experimental Research and Medical Studies, University of Torino, Torino, Italy
| | - Elisabetta Mereu
- Department of Molecular Biotechnology and Health Sciences; Center for Experimental Research and Medical Studies, University of Torino, Torino, Italy
| | - Giorgio Inghirami
- Department of Molecular Biotechnology and Health Sciences; Center for Experimental Research and Medical Studies, University of Torino, Torino, Italy; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Roberto Piva
- Department of Molecular Biotechnology and Health Sciences; Center for Experimental Research and Medical Studies, University of Torino, Torino, Italy.
| |
Collapse
|
117
|
Tripathi S, Mathur S, Deshmukh P, Manjula R, Padmanabhan B. A Novel Phenanthridionone Based Scaffold As a Potential Inhibitor of the BRD2 Bromodomain: Crystal Structure of the Complex. PLoS One 2016; 11:e0156344. [PMID: 27243809 PMCID: PMC4886958 DOI: 10.1371/journal.pone.0156344] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 05/12/2016] [Indexed: 11/18/2022] Open
Abstract
Bromodomain containing proteins recognize the level of histone acetylation and regulate epigenetically controlled processes like gene transcription and chromatin modification. The BET (bromodomain and extra-terminal) family proteins, which are transcriptional co-regulators, have been implicated in the pathogenesis of cancer, neurodegenerative disorders, and defects in embryonic stem cell differentiation. Inhibitors selectively targeting the BET bromodomains can pave the path for new drug discovery against several forms of major diseases. By a rational structure-based approach, we have identified a new inhibitor (NSC127133) of the second bromodomain (BD2) of the BET family protein BRD2 using the NCI Diversity Set III library. A high-resolution crystal structure of the BRD2-BD2 in complex with this compound and in apo- form is refined to 0.91 and 0.94 Å, respectively. The compound, which is a phenanthridinone derivative, binds well to the acetyl-lysine binding pocket of BD2 and displays significant hydrophobic and hydrophilic interactions. Moreover, the atomic resolution data obtained in this study allowed us to visualize certain structural features of BD2 which remained unobserved so far. We propose that the discovered compound may be a potential molecule to develop a new library for inhibiting the BRD2-BD2 function.
Collapse
Affiliation(s)
- Shailesh Tripathi
- Department of Biophysics, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bangalore, 560029, India
| | - Shruti Mathur
- Department of Biophysics, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bangalore, 560029, India
| | - Prashant Deshmukh
- Department of Biophysics, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bangalore, 560029, India
| | - Ramu Manjula
- Department of Biophysics, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bangalore, 560029, India
| | - Balasundaram Padmanabhan
- Department of Biophysics, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bangalore, 560029, India
- * E-mail: ;
| |
Collapse
|
118
|
Zahid MF, Patnaik MM, Gangat N, Hashmi SK, Rizzieri DA. Insight into the molecular pathophysiology of myelodysplastic syndromes: targets for novel therapy. Eur J Haematol 2016; 97:313-20. [DOI: 10.1111/ejh.12771] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2016] [Indexed: 01/07/2023]
Affiliation(s)
| | - Mrinal M. Patnaik
- Division of Hematology; Mayo Clinic; Rochester MN USA
- Mayo Clinic Transplant Center; Blood and Marrow Transplant Program; Mayo Clinic; Rochester MN USA
| | | | - Shahrukh K. Hashmi
- Division of Hematology; Mayo Clinic; Rochester MN USA
- Mayo Clinic Transplant Center; Blood and Marrow Transplant Program; Mayo Clinic; Rochester MN USA
| | - David A. Rizzieri
- Division of Hematologic Malignancies & Cellular Therapy; Duke University Medical Center; Durham NC USA
| |
Collapse
|
119
|
Abstract
BET proteins have recently become recognized for their role in a broad range of cancers and are defined by the presence of two acetyl-histone reading bromodomains and an ET domain. This family of proteins includes BRD2, BRD3, BRD4, and BRDT. BRD4 is the most-studied BET protein in cancer, and normally serves as an epigenetic reader that links active chromatin marks to transcriptional elongation through activation of RNA polymerase II. The role of BRD3 and BRD4 first became known in cancer as mutant oncoproteins fused to the p300-recruiting NUT protein in a rare aggressive subtype of squamous cell cancer known as NUT midline carcinoma (NMC). BET inhibitors are acetyl-histone mimetics that specifically bind BET bromodomains, competitively inhibiting its engagement with chromatin. The antineoplastic effects of BET inhibitors were first demonstrated in NMC and have since been shown to be effective at inhibiting the growth of many different cancers, particularly acute leukemia. BET inhibitors have also been instrumental as tool compounds that have demonstrated the key role of BRD4 in driving NMC and non-NMC cancer growth. Many clinical trials enrolling patients with hematologic and solid tumors are ongoing, with encouraging preliminary findings. BET proteins BRD2, BRD3, and BRD4 are expressed in nearly all cells of the body, so there are concerns of toxicity with BET inhibitors, as well as the development of resistance. Toxicity and resistance may be overcome by combining BET inhibitors with other targeted inhibitors, or through the use of novel BET inhibitor derivatives.
Collapse
Affiliation(s)
- C A French
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
120
|
Wadhwa E, Nicolaides T. Bromodomain Inhibitor Review: Bromodomain and Extra-terminal Family Protein Inhibitors as a Potential New Therapy in Central Nervous System Tumors. Cureus 2016; 8:e620. [PMID: 27382528 PMCID: PMC4917374 DOI: 10.7759/cureus.620] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The bromodomain and extraterminal (BET) family proteins associate with transcriptional activation through interaction with acetylated chromatin, therefore playing a key role as epigenetic regulators. BET proteins serve to regulate the expression of importance oncogenes, including those involved in apoptosis as well as cell cycle progression. Due to this potential as an epigenetic target, small molecule inhibition of BET proteins have been investigated and demonstrate promising activity in both solid and hematologic malignancies, including brain tumors. Glioblastoma multiforme (GBM), subsets of medulloblastoma, and diffuse intrinsic pontine glioma (DIPG) are types of brain tumors with dismal prognoses, and as such have been the subjects of preclinical studies using BET inhibitors both in vivo and in vitro. While results from these preclinical investigations have shown promise, clinical trials are in early phases at this time. In this review, we will summarize the current literature on BET family proteins, their potential as therapeutic targets in brain tumors as well as other malignancies, and the preclinical and clinical investigations that have been undertaken to date.
Collapse
Affiliation(s)
| | - Theodore Nicolaides
- Neurological Surgery and Pediatrics, University of California, San Francisco
| |
Collapse
|
121
|
Palmer WS. Development of small molecule inhibitors of BRPF1 and TRIM24 bromodomains. DRUG DISCOVERY TODAY. TECHNOLOGIES 2016; 19:65-71. [PMID: 27769360 DOI: 10.1016/j.ddtec.2016.06.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/27/2016] [Accepted: 06/03/2016] [Indexed: 06/06/2023]
Abstract
The entry of small molecule inhibitors of the bromodomain and extra C-terminal domain (BET) family of bromodomains into the clinic has demonstrated the therapeutic potential for this class of epigenetic acetyl-lysine reader proteins. Within the past two years, the development of potent inhibitors for the bromodomain and PHD finger containing protein (BRPF) family and the tripartite motif containing protein 24 (TRIM24) have been reported and are the subject of this review. Both proteins contain other domains with diverse functions and can also be part of a complex of proteins which have implications in epigenetic signaling and disease. These new small molecule tools will be useful for unraveling the biological contribution of the bromodomain and enable pharmacological validation of these proteins.
Collapse
Affiliation(s)
- Wylie S Palmer
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| |
Collapse
|
122
|
Bid HK, Phelps DA, Xaio L, Guttridge DC, Lin J, London C, Baker LH, Mo X, Houghton PJ. The Bromodomain BET Inhibitor JQ1 Suppresses Tumor Angiogenesis in Models of Childhood Sarcoma. Mol Cancer Ther 2016; 15:1018-28. [PMID: 26908627 DOI: 10.1158/1535-7163.mct-15-0567] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 02/04/2016] [Indexed: 02/04/2023]
Abstract
The bromodomain and extra-terminal domain inhibitor JQ1 has marked antitumor activity against several hematologic malignancies as well as solid tumor models. Here, we investigated its activity in vitro and in vivo against models of childhood rhabdomyosarcoma and Ewing sarcoma. In vitro, JQ1 (but not the inactive enantiomer JQ1R) inhibited cell proliferation and increased G1 fraction of cells, although there was no correlation between cell line sensitivity and suppression of c-MYC or MYCN. In vivo, xenografts showed significant inhibition of growth during the period of treatment, and rapid regrowth after treatment was stopped, activity typical of antiangiogenic agents. Furthermore, xenografts derived from cell lines intrinsically resistant or sensitive to JQ1 in vitro had similar sensitivity in vivo as xenografts. Further investigation showed that JQ1 reduced tumor vascularization. This was secondary to both drug-induced downregulation of tumor-derived growth factors and direct effects of JQ1 on vascular elements. JQ1 suppressed VEGF-stimulated vascularization of Matrigel plugs in mice, and in vitro suppressed differentiation, proliferation, and invasion of human umbilical cord vascular endothelial cells (HUVEC). In HUVECs, JQ1 partially suppressed c-MYC levels, but dramatically reduced AP-1 levels and activity through suppression of the AP-1-associated protein FOSL1. Our data suggest that the antitumor activity of JQ1 in these sarcoma models is largely a consequence of its antiangiogenic activity. Mol Cancer Ther; 15(5); 1018-28. ©2016 AACR.
Collapse
Affiliation(s)
- Hemant K Bid
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio
| | - Doris A Phelps
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio
| | - Linlin Xaio
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio
| | - Denis C Guttridge
- Center for Regenerative Medicine, Ohio State University, Columbus, Ohio
| | - Jiayuh Lin
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio
| | - Cheryl London
- College of Veterinary Medicine, Ohio State University, Columbus, Ohio
| | - Laurence H Baker
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Xiaokui Mo
- Center for Biostatistics, Ohio State University, Columbus, Ohio
| | - Peter J Houghton
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio. Greehey Children's Cancer Research Institute, San Antonio, Texas.
| |
Collapse
|
123
|
Albrecht BK, Gehling VS, Hewitt MC, Vaswani RG, Côté A, Leblanc Y, Nasveschuk CG, Bellon S, Bergeron L, Campbell R, Cantone N, Cooper MR, Cummings RT, Jayaram H, Joshi S, Mertz JA, Neiss A, Normant E, O’Meara M, Pardo E, Poy F, Sandy P, Supko J, Sims RJ, Harmange JC, Taylor AM, Audia JE. Identification of a Benzoisoxazoloazepine Inhibitor (CPI-0610) of the Bromodomain and Extra-Terminal (BET) Family as a Candidate for Human Clinical Trials. J Med Chem 2016; 59:1330-9. [DOI: 10.1021/acs.jmedchem.5b01882] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Brian K. Albrecht
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Victor S. Gehling
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Michael C. Hewitt
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Rishi G. Vaswani
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Alexandre Côté
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Yves Leblanc
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Christopher G. Nasveschuk
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Steve Bellon
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Louise Bergeron
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Robert Campbell
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Nico Cantone
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Michael R. Cooper
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Richard T. Cummings
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Hariharan Jayaram
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Shivangi Joshi
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Jennifer A. Mertz
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Adrianne Neiss
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Emmanuel Normant
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Michael O’Meara
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Eneida Pardo
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Florence Poy
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Peter Sandy
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Jeffrey Supko
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Robert J. Sims
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Jean-Christophe Harmange
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Alexander M. Taylor
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - James E. Audia
- Constellation Pharmaceuticals, 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
124
|
Dubois S, Jardin F. The role of next-generation sequencing in understanding the genomic basis of diffuse large B cell lymphoma and advancing targeted therapies. Expert Rev Hematol 2016; 9:255-69. [PMID: 26652775 DOI: 10.1586/17474086.2016.1130616] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Next Generation Sequencing (NGS) has redefined the genetic landscape of Diffuse Large B-Cell Lymphoma (DLBCL) by identifying recurrent somatic mutations. Importantly, in some cases these mutations impact potentially actionable targets, thus affording novel personalized therapy opportunities. At the forefront of today's precision therapy era, how to best incorporate NGS into daily clinical practice is of primordial concern, in order to tailor patient's treatment regimens according to their individual mutational profiles. With the advent of cell-free DNA sequencing, which provides a sensitive and less invasive means of monitoring DLBCL patients, the clinical feasibility of NGS has been greatly improved. This article reviews the current landscape of DLBCL mutations, as well as the targeted therapies developed to counter their effects, and discusses how best to utilize NGS data for treatment decision-making.
Collapse
Affiliation(s)
- Sydney Dubois
- a Inserm U918, Centre Henri Becquerel , Université de Rouen, IRIB , Rouen , France
| | - Fabrice Jardin
- a Inserm U918, Centre Henri Becquerel , Université de Rouen, IRIB , Rouen , France.,b Department of Hematology , Centre Henri Becquerel , Rouen , France
| |
Collapse
|
125
|
Ferri E, Petosa C, McKenna CE. Bromodomains: Structure, function and pharmacology of inhibition. Biochem Pharmacol 2015; 106:1-18. [PMID: 26707800 DOI: 10.1016/j.bcp.2015.12.005] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/08/2015] [Indexed: 12/22/2022]
Abstract
Bromodomains are epigenetic readers of histone acetylation involved in chromatin remodeling and transcriptional regulation. The human proteome comprises 46 bromodomain-containing proteins with a total of 61 bromodomains, which, despite highly conserved structural features, recognize a wide array of natural peptide ligands. Over the past five years, bromodomains have attracted great interest as promising new epigenetic targets for diverse human diseases, including inflammation, cancer, and cardiovascular disease. The demonstration in 2010 that two small molecule compounds, JQ1 and I-BET762, potently inhibit proteins of the bromodomain and extra-terminal (BET) family with translational potential for cancer and inflammatory disease sparked intense efforts in academia and pharmaceutical industry to develop novel bromodomain antagonists for therapeutic applications. Several BET inhibitors are already in clinical trials for hematological malignancies, solid tumors and cardiovascular disease. Currently, the field faces the challenge of single-target selectivity, especially within the BET family, and of overcoming problems related to the development of drug resistance. At the same time, new trends in bromodomain inhibitor research are emerging, including an increased interest in non-BET bromodomains and a focus on drug synergy with established antitumor agents to improve chemotherapeutic efficacy. This review presents an updated view of the structure and function of bromodomains, traces the development of bromodomain inhibitors and their potential therapeutic applications, and surveys the current challenges and future directions of this vibrant new field in drug discovery.
Collapse
Affiliation(s)
- Elena Ferri
- Department of Chemistry, Dana and David Dornsife College of Letters, Arts and Sciences, University of Southern California, University Park Campus, Los Angeles, CA 90089, United States
| | - Carlo Petosa
- Université Grenoble Alpes, Institut de Biologie Structurale (IBS), 71 Avenue des Martyrs, 38044 Grenoble, France; Centre National de la Recherche Scientifique, IBS, 38044 Grenoble, France; Commissariat à l'Energie Atomique et aux Energies Alternatives, IBS, 38044 Grenoble, France
| | - Charles E McKenna
- Department of Chemistry, Dana and David Dornsife College of Letters, Arts and Sciences, University of Southern California, University Park Campus, Los Angeles, CA 90089, United States.
| |
Collapse
|
126
|
Hagenbuchner J, Ausserlechner MJ. Targeting transcription factors by small compounds--Current strategies and future implications. Biochem Pharmacol 2015; 107:1-13. [PMID: 26686579 DOI: 10.1016/j.bcp.2015.12.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 12/08/2015] [Indexed: 12/16/2022]
Abstract
Transcription factors are central regulators of gene expression and critically steer development, differentiation and death. Except for ligand-activated nuclear receptors, direct modulation of transcription factor function by small molecules is still widely regarded as "impossible". This "un-druggability" of non-ligand transcription factors is due to the fact that the interacting surface between transcription factor and DNA is huge and subject to significant changes during DNA-binding. Besides some "success studies" with compounds that directly interfere with DNA binding, drug targeting approaches mostly address protein-protein interfaces with essential co-factors, transcription factor dimerization partners, chaperone proteins or proteins that regulate subcellular shuttling. An alternative strategy represent DNA-intercalating, alkylating or DNA-groove-binding compounds that either block transcription factor-binding or change the 3D-conformation of the consensus DNA-strand. Recently, much interest has been focused on chromatin reader proteins that steer the recruitment and activity of transcription factors to a gene transcription start site. Several small compounds demonstrate that these epigenetic reader proteins are exciting new drug targets for inhibiting lineage-specific transcription in cancer therapy. In this research update we will discuss recent advances in targeting transcription factors with small compounds, the challenges that are related to the complex function and regulation of these proteins and also the possible future directions and applications of transcription factor drug targeting.
Collapse
Affiliation(s)
- Judith Hagenbuchner
- Department of Pediatrics II, Medical University Innsbruck, Innrain 66, A-6020 Innsbruck, Austria
| | - Michael J Ausserlechner
- Department of Pediatrics I, Medical University Innsbruck, Innrain 66, A-6020 Innsbruck, Austria.
| |
Collapse
|
127
|
Romero FA, Taylor AM, Crawford TD, Tsui V, Côté A, Magnuson S. Disrupting Acetyl-Lysine Recognition: Progress in the Development of Bromodomain Inhibitors. J Med Chem 2015; 59:1271-98. [PMID: 26572217 DOI: 10.1021/acs.jmedchem.5b01514] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bromodomains, small protein modules that recognize acetylated lysine on histones, play a significant role in the epigenome, where they function as "readers" that ultimately determine the functional outcome of the post-translational modification. Because the initial discovery of selective BET inhibitors have helped define the role of that protein family in oncology and inflammation, BET bromodomains have continued to garner the most attention of any other bromodomain. More recently, non-BET bromodomain inhibitors that are potent and selective have been disclosed for ATAD2, CBP, BRD7/9, BRPF, BRPF/TRIM24, CECR2, SMARCA4, and BAZ2A/B. Such novel inhibitors can be used to probe the physiological function of these non-BET bromodomains and further understanding of their role in certain disease states. Here, we provide an update to the progress in identifying selective bromodomain inhibitors and their use as biological tools, as well as our perspective on the field.
Collapse
Affiliation(s)
- F Anthony Romero
- Discovery Chemistry, Genentech, Inc. 1 DNA Way, South San Francisco, California 94080, United States
| | - Alexander M Taylor
- Constellation Pharmaceuticals, Inc. 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Terry D Crawford
- Discovery Chemistry, Genentech, Inc. 1 DNA Way, South San Francisco, California 94080, United States
| | - Vickie Tsui
- Discovery Chemistry, Genentech, Inc. 1 DNA Way, South San Francisco, California 94080, United States
| | - Alexandre Côté
- Constellation Pharmaceuticals, Inc. 215 First Street, Suite 200, Cambridge, Massachusetts 02142, United States
| | - Steven Magnuson
- Discovery Chemistry, Genentech, Inc. 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
128
|
Cai Q, Medeiros LJ, Xu X, Young KH. MYC-driven aggressive B-cell lymphomas: biology, entity, differential diagnosis and clinical management. Oncotarget 2015; 6:38591-38616. [PMID: 26416427 PMCID: PMC4770723 DOI: 10.18632/oncotarget.5774] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 09/04/2015] [Indexed: 01/09/2023] Open
Abstract
MYC, a potent oncogene located at chromosome locus 8q24.21, was identified initially by its involvement in Burkitt lymphoma with t(8;14)(q24;q32). MYC encodes a helix-loop-helix transcription factor that accentuates many cellular functions including proliferation, growth and apoptosis. MYC alterations also have been identified in other mature B-cell neoplasms and are associated with aggressive clinical behavior. There are several regulatory factors and dysregulated signaling that lead to MYC up-regulation in B-cell lymphomas. One typical example is the failure of physiological repressors such as Bcl6 or BLIMP1 to suppress MYC over-expression. In addition, MYC alterations are often developed concurrently with other genetic alterations that counteract the proapoptotic function of MYC. In this review, we discuss the physiologic function of MYC and the role that MYC likely plays in the pathogenesis of B-cell lymphomas. We also summarize the role MYC plays in the diagnosis, prognostication and various strategies to detect MYC rearrangement and expression.
Collapse
Affiliation(s)
- Qingqing Cai
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - L. Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xiaolu Xu
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Ken H. Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The University of Texas School of Medicine, Graduate School of Biomedical Sciences, Houston, Texas, USA
| |
Collapse
|
129
|
Benedetti R, Conte M, Iside C, Altucci L. Epigenetic-based therapy: From single- to multi-target approaches. Int J Biochem Cell Biol 2015; 69:121-31. [PMID: 26494003 DOI: 10.1016/j.biocel.2015.10.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 10/13/2015] [Accepted: 10/14/2015] [Indexed: 12/20/2022]
Abstract
The treatment of cancer has traditionally been based on the identification of a single molecule and/or enzymatic function (target) responsible for a particular phenotype, and therefore on the ability to stimulate, attenuate or inhibit its activity through the use of selective compounds. However, cancer is no longer considered a disease caused by a single factor, but is now recognized as a multi-factorial disorder. Genetic, epigenetic and metabolic factors all contribute to neoplasia, causing significant changes in molecular networks that govern cell growth, development, death and specialization. Consequently, many antitumor therapies are no longer directed against a single target but the biological system as a whole, in which functions determining the onset and maintenance of a physio-pathological state are modulated. The field of epi-drug discovery is currently in a transitional phase where the search for putative anticancer drugs is shifting from single-target-oriented molecules to network-active compounds and to epi-drugs used in combination with other epi-agents and with traditional chemotherapeutics. This review illustrates the pros and cons of each therapeutic option, providing examples in support of single-target and multi (network)-target epi-drug approaches.
Collapse
Affiliation(s)
- Rosaria Benedetti
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università di Napoli, Vico L. De Crecchio 7, 80138 Napoli, Italy.
| | - Mariarosaria Conte
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università di Napoli, Vico L. De Crecchio 7, 80138 Napoli, Italy
| | - Concetta Iside
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università di Napoli, Vico L. De Crecchio 7, 80138 Napoli, Italy
| | - Lucia Altucci
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università di Napoli, Vico L. De Crecchio 7, 80138 Napoli, Italy; Istituto di Genetica e Biofisica, Adriano Buzzati Traverso, CNR-IGB, Via P. Castellino 111, 80131 Napoli, Italy.
| |
Collapse
|
130
|
Abstract
Cancer genome sequencing has created an opportunity for precision medicine. Thus far, genetic alterations can only be used to guide treatment for small subsets of certain cancer types with these key alterations. Similar to mutations, epigenetic events are equally suitable for personalized medicine. DNA methylation alterations have been used to identify tumor-specific drug responsive markers. Methylation of MGMT sensitizes gliomas to alkylating agents is an example of epigenetic personalized medicine. Recent studies have revealed that 5-azacytidine and decitabine show activity in myelodysplasia, lung and other cancers. There are currently at least 20 kinds of histone deacetylase inhibitors in clinical testing. Inhibitors targeting other epigenetic regulators are being clinically tested, such as EZH2 inhibitor EPZ-6438.
Collapse
Affiliation(s)
- Wenji Yan
- Department of Gastroenterology & Hepatology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing 100853, China
| | - James G Herman
- The Hillman Cancer Center, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Suite 2.18/Research, Pittsburgh, PA 15213, USA
| | - Mingzhou Guo
- Department of Gastroenterology & Hepatology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing 100853, China
| |
Collapse
|