MEK Inhibitors Overcome Resistance to BET Inhibition Across a Number of Solid and Hematologic Cancers Anastasia Wyce1, Jeanne J

Total Page:16

File Type:pdf, Size:1020Kb

MEK Inhibitors Overcome Resistance to BET Inhibition Across a Number of Solid and Hematologic Cancers Anastasia Wyce1, Jeanne J Wyce et al. Oncogenesis (2018) 7:35 DOI 10.1038/s41389-018-0043-9 Oncogenesis ARTICLE Open Access MEK inhibitors overcome resistance to BET inhibition across a number of solid and hematologic cancers Anastasia Wyce1, Jeanne J. Matteo1,ShawnW.Foley1, Daniel J. Felitsky2, Satyajit R. Rajapurkar1, Xi-Ping Zhang1, Melissa C. Musso1,SusanKorenchuk1, Natalie O. Karpinich1, Kathryn M. Keenan1, Melissa Stern1, Lijoy K. Mathew1, Charles F. McHugh1, Michael T. McCabe1, Peter J. Tummino1,3,RyanG.Kruger1, Christopher Carpenter1 and Olena Barbash1 Abstract BET inhibitors exhibit broad activity in cancer models, making predictive biomarkers challenging to define. Here we investigate the biomarkers of activity of the clinical BET inhibitor GSK525762 (I-BET; I-BET762) across cancer cell lines and demonstrate that KRAS mutations are novel resistance biomarkers. This finding led us to combine BET with RAS pathway inhibition using MEK inhibitors to overcome resistance, which resulted in synergistic effects on growth and survival in RAS pathway mutant models as well as a subset of cell lines lacking RAS pathway mutations. GSK525762 treatment up-regulated p-ERK1/2 levels in both RAS pathway wild-type and mutant cell lines, suggesting that MEK/ERK pathway activation may also be a mechanism of adaptive BET inhibitor resistance. Importantly, gene expression studies demonstrated that the BET/MEK combination uniquely sustains down-regulation of genes associated with mitosis, leading to prolonged growth arrest that is not observed with either single agent therapy. These studies 1234567890():,; 1234567890():,; highlight a potential to enhance the clinical benefit of BET and MEK inhibitors and provide a strong rationale for clinical evaluation of BET/MEK combination therapies in cancer. Introduction inhibitors including transcriptional suppression of onco- BET proteins (BRD2, BRD3, BRD4, and BRDT) mod- genes3, there is no consensus and it is likely that ulate expression of genes involved in cell growth and mechanisms vary, thus making the identification of pre- oncogenesis by binding to acetylated chromatin via their dictive biomarkers difficult. bromodomains, which in turn recruit downstream effec- Although BET inhibitors show broad activity in many tors that promote transcription. Selective BET inhibitors, cancer types, within each there are resistant models. such as I-BET151 and the clinical molecule GSK525762 Understanding the basis of BET inhibitor sensitivity and (I-BET762; I-BET)1,2, abrogate binding of BET proteins to resistance is important to inform the clinical development acetylated chromatin thereby inhibiting BET-dependent of BET inhibitors as monotherapies and to identify transcription1,2. BET inhibitors exhibit broad anti- rational combinations. To this end, we analyzed genetic proliferative activity in cancer models3. Although several data from a large set of cell lines treated with GSK525762 mechanisms have been implicated in the efficacy of BET to identify biomarkers of sensitivity and resistance. From these studies, we identified KRAS mutations as a sig- nificant predictor of resistance to BET inhibition. This led Correspondence: Olena Barbash ([email protected]) us to hypothesize that combinations with inhibitors of 1 Cancer Epigenetics DPU, Oncology R&D, GlaxoSmithKline, Collegeville, PA, RAS signaling, such as MEK inhibitors, may further USA 2Target Sciences, GlaxoSmithKline, Upper Merion, PA, USA improve upon BET inhibitor efficacy. Indeed, we observed Full list of author information is available at the end of the article © The Author(s) 2018 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a linktotheCreativeCommons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/. Oncogenesis Wyce et al. Oncogenesis (2018) 7:35 Page 2 of 12 broad synergistic effects for BET/MEK combinations inhibitor combinations in cancer cell lines with and across cancer models, which we attribute to profound and without RAS pathway mutations (mutations in RAS, sustained inhibition of MEK/ERK signaling that is speci- BRAF, or NF1), and with varying levels of sensitivity to fically observed with the combination ultimately leading GSK525762. For these studies, we utilized the BET inhi- to growth arrest and cell death. bitor I-BET151, (pharmacological profile highly similar to GSK5257621; Supplemental Figure S3), and the clinical Results MEK inhibitor PD0325901. Combination of I-BET151 RAS mutations are novel biomarkers of resistance to and PD0325901 produced synergistic effects (CI < 0.78; GSK525762 see Methods) most commonly in, but not limited to, RAS To identify genetic predictors of sensitivity or resistance pathway mutant models. Synergy was observed in multi- to BET inhibitors we first examined the anti-proliferative ple tumor types (Fig. 2a, Supplemental Table S3), with the activity of GSK525762 in ~230 cancer cell lines. Hema- combination improving growth inhibition (gIC50, gIC100) tologic cancer cell lines were highly sensitive (low growth and/or cytotoxicity (dEC50) even in cell lines sensitive to IC50 values and net cell death) to GSK525762, whereas single agent BET inhibition (Fig. 2b, c, Supplemental solid tumor models exhibited a wide range in sensitivity Table S1, S3). Across all cell lines evaluated, there was a (gIC50 13 nM to > 29.3 µM; partial, cytostatic, and cyto- significant correlation (p = 0.022; Fisher’s exact test) toxic responses), providing an opportunity to compare the between RAS pathway mutations and synergy (Fig. 2a; genetic profiles of a large number of sensitive and resis- Supplemental Table S3). However, synergy was not tant lines (Fig. 1a, b, Supplemental Table S1). Using restricted to mutant models, suggesting that additional publicly available data for 19,312 genes with protein- mechanisms may drive combination effects. changing mutations, we performed unbiased analyses of Given the reported variability in biological activity of genetic predictors of sensitivity or resistance to MEK inhibitors based on differing mechanisms of inhi- 4 GSK525762 based on gIC50 values. These analyses iden- bition , we evaluated combinations with additional MEK tified 634 genes with protein-changing mutations that inhibitors in cell lines that exhibited synergistic responses correspond to resistance or sensitivity to GSK525762 to I-BET151 and PD0325901. Synergy was observed with (Wilcoxon rank sum test p < 0.05; Supplemental Table S2, a variety of structurally distinct MEK inhibitors, including Supplemental Figure S1). Notably, KRAS mutations GDC-0623, trametinib, and cobimetinib (Supplemental were among the top five most significant (p = 0.0001) Figs. S4A-4B). Importantly, combination of the clinical associations, with a clear enrichment of KRAS mutations BET inhibitor GSK525762 with therapeutic concentra- 5 among the more resistant cell lines (median gIC50 tions of trametinib resulted in decreased cell viability KRAS mutant = 1667 nM vs WT = 550 nM; Fig. 1c). (Supplemental Figure S4C). Analysis of mutations at the amino acid level further To determine if in vitro synergy translates into identified KRAS G12 missense mutations as significantly improved in vivo efficacy, we profiled the activity of the (p = 0.00006) associated with resistance (Supplemental BET/MEK combination in xenografts representing var- Figure S2). A similar association (p = 0.004) between ious tumor types and RAS pathway mutations. Combi- KRAS mutations and resistance was observed in colorectal nation of GSK525762 and trametinib significantly delayed cancer (CRC) cell lines, where KRAS mutations are fre- tumor growth in xenograft models of CRC (RKO; quent (mutations in 13/22 lines; 59%) (Fig. 1d), indicating BRAFmut, NF1mut), triple-negative breast cancer (TNBC; that the association with resistance is not driven by the MDA-MB-231; KRASmut, BRAFmut, NF1mut), and multi- general responsiveness of individual tumor types. Finally, ple myeloma (MM; RPMI-8226; KRASmut) (Fig. 2d; Sup- KRAS mutations were significantly depleted (p = 0.003; plemental Figures S5-S7), and significantly improved Fisher’s exact test) among the cell lines exhibiting net cell tumor growth inhibition in a xenograft model of pan- death in response to GSK525762 (Supplemental Table creatic cancer (PaCa; HPAF-II; KRASmut) (Supplemental S1), suggesting that activated RAS signaling may be a Figure S8) at well-tolerated doses (Supplemental Figure mechanism by which cancer cells are able to survive BET S9). inhibition. Synergy for BET/MEK combinations in RAS pathway wild Broad synergy for combinations of BET and MEK inhibitors type cell lines in RAS pathway mutant cancer cell lines Although RAS pathway mutations predict resistance to The strong association between KRAS mutations and BET inhibitors and sensitivity to BET/MEK combinations, resistance to GSK525762 suggested that agents targeting we also observed synergy
Recommended publications
  • Functional Roles of Bromodomain Proteins in Cancer
    cancers Review Functional Roles of Bromodomain Proteins in Cancer Samuel P. Boyson 1,2, Cong Gao 3, Kathleen Quinn 2,3, Joseph Boyd 3, Hana Paculova 3 , Seth Frietze 3,4,* and Karen C. Glass 1,2,4,* 1 Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, Colchester, VT 05446, USA; [email protected] 2 Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA; [email protected] 3 Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05405, USA; [email protected] (C.G.); [email protected] (J.B.); [email protected] (H.P.) 4 University of Vermont Cancer Center, Burlington, VT 05405, USA * Correspondence: [email protected] (S.F.); [email protected] (K.C.G.) Simple Summary: This review provides an in depth analysis of the role of bromodomain-containing proteins in cancer development. As readers of acetylated lysine on nucleosomal histones, bromod- omain proteins are poised to activate gene expression, and often promote cancer progression. We examined changes in gene expression patterns that are observed in bromodomain-containing proteins and associated with specific cancer types. We also mapped the protein–protein interaction network for the human bromodomain-containing proteins, discuss the cellular roles of these epigenetic regu- lators as part of nine different functional groups, and identify bromodomain-specific mechanisms in cancer development. Lastly, we summarize emerging strategies to target bromodomain proteins in cancer therapy, including those that may be essential for overcoming resistance. Overall, this review provides a timely discussion of the different mechanisms of bromodomain-containing pro- Citation: Boyson, S.P.; Gao, C.; teins in cancer, and an updated assessment of their utility as a therapeutic target for a variety of Quinn, K.; Boyd, J.; Paculova, H.; cancer subtypes.
    [Show full text]
  • Bromodomain Protein BRDT Directs ΔNp63 Function and Super
    Cell Death & Differentiation (2021) 28:2207–2220 https://doi.org/10.1038/s41418-021-00751-w ARTICLE Bromodomain protein BRDT directs ΔNp63 function and super-enhancer activity in a subset of esophageal squamous cell carcinomas 1 1 2 1 1 2 Xin Wang ● Ana P. Kutschat ● Moyuru Yamada ● Evangelos Prokakis ● Patricia Böttcher ● Koji Tanaka ● 2 3 1,3 Yuichiro Doki ● Feda H. Hamdan ● Steven A. Johnsen Received: 25 August 2020 / Revised: 3 February 2021 / Accepted: 4 February 2021 / Published online: 3 March 2021 © The Author(s) 2021. This article is published with open access Abstract Esophageal squamous cell carcinoma (ESCC) is the predominant subtype of esophageal cancer with a particularly high prevalence in certain geographical regions and a poor prognosis with a 5-year survival rate of 15–25%. Despite numerous studies characterizing the genetic and transcriptomic landscape of ESCC, there are currently no effective targeted therapies. In this study, we used an unbiased screening approach to uncover novel molecular precision oncology targets for ESCC and identified the bromodomain and extraterminal (BET) family member bromodomain testis-specific protein (BRDT) to be 1234567890();,: 1234567890();,: uniquely expressed in a subgroup of ESCC. Experimental studies revealed that BRDT expression promotes migration but is dispensable for cell proliferation. Further mechanistic insight was gained through transcriptome analyses, which revealed that BRDT controls the expression of a subset of ΔNp63 target genes. Epigenome and genome-wide occupancy studies, combined with genome-wide chromatin interaction studies, revealed that BRDT colocalizes and interacts with ΔNp63 to drive a unique transcriptional program and modulate cell phenotype. Our data demonstrate that these genomic regions are enriched for super-enhancers that loop to critical ΔNp63 target genes related to the squamous phenotype such as KRT14, FAT2, and PTHLH.
    [Show full text]
  • Overcoming BET Inhibitor Resistance in Malignant Peripheral Nerve Sheath Tumors
    Author Manuscript Published OnlineFirst on February 22, 2019; DOI: 10.1158/1078-0432.CCR-18-2437 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Cooper and Patel, et al. Page 1 Overcoming BET inhibitor resistance in malignant peripheral nerve sheath tumors Jonathan M. Cooper1,6, Amish J. Patel1,2,6, Zhiguo Chen1,7, Chung-Ping Liao1,7, Kun Chen1,7, Juan Mo1, Yong Wang1, Lu Q. Le1,3,4,5 1Department of Dermatology, 2Cancer Biology Graduate Program, 3Simmons Comprehensive Cancer Center, 4UTSW Comprehensive Neurofibromatosis Clinic, 5Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, 75390-9133, USA 6Co-first authors. 7These authors contributed equally. Author for correspondence: Lu Q. Le, M.D., Ph.D. Associate Professor Department of Dermatology Simmons Comprehensive Cancer Center UT Southwestern Medical Center Phone: (214) 648-5781 Fax: (214) 648-5553 E-mail: [email protected] Running Title: Cancer cell sensitivity and resistance to BET inhibitors Keywords: Neurofibromatosis Type1, NF1, Malignant Peripheral Nerve Sheath Tumor, MPNST, Neurofibrosarcomas, JQ1, BRD4 levels, BET inhibitor sensitivity and resistance, BET Bromodomain, PROTAC, ARV771 The authors have declared that no conflict of interest exists. Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 22, 2019; DOI: 10.1158/1078-0432.CCR-18-2437 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Cooper and Patel, et al. Page 2 Statement of Translational Relevance Malignant peripheral nerve sheath tumors (MPNST) are aggressive sarcomas with no effective therapies.
    [Show full text]
  • Targeting MYC Dependence in Cancer by Inhibiting BET Bromodomains
    Targeting MYC dependence in cancer by inhibiting BET bromodomains Jennifer A. Mertz1, Andrew R. Conery1, Barbara M. Bryant, Peter Sandy, Srividya Balasubramanian, Deanna A. Mele, Louise Bergeron, and Robert J. Sims III2 Constellation Pharmaceuticals, Inc., Cambridge, MA 02142 Edited* by Robert N. Eisenman, Fred Hutchinson Cancer Research Center, Seattle, WA, and approved August 30, 2011 (received for review May 23, 2011) The MYC transcription factor is a master regulator of diverse in preclinical animal models of endotoxic shock caused by acti- cellular functions and has been long considered a compelling vation of Toll-like receptors that trigger the up-regulation of therapeutic target because of its role in a range of human cytokines controlled by NF-κB (7). Importantly, although RNAi- malignancies. However, pharmacologic inhibition of MYC function mediated depletion of any one individual BET protein has broad has proven challenging because of both the diverse mechanisms effects on transcription, pharmacologic inhibition of the BET driving its aberrant expression and the challenge of disrupting bromodomains perturbs the transcription of a narrower range protein–DNA interactions. Here, we demonstrate the rapid and po- of genes (7). These findings suggest an opportunity to use BET- tent abrogation of MYC gene transcription by representative small bromodomain inhibitors to fine-tune key regulatory pathways for molecule inhibitors of the BET family of chromatin adaptors. MYC therapeutic benefit. transcriptional suppression was observed in the context of the nat- BET inhibitors exhibit anticancer effects in both in vitro and ural, chromosomally translocated, and amplified gene locus. Inhi- in vivo models of midline carcinoma (8), a rare but lethal disease bition of BET bromodomain–promoter interactions and subsequent that frequently harbors a chromosomal translocation in which reduction of MYC transcript and protein levels resulted in G1 arrest the bromodomains of BRD4 are fused to the NUT gene (9).
    [Show full text]
  • Targeting Transcriptional Regulation of SARS-Cov-2 Entry Factors ACE2 and TMPRSS2
    Targeting transcriptional regulation of SARS-CoV-2 entry factors ACE2 and TMPRSS2 Yuanyuan Qiaoa,b,c,1, Xiao-Ming Wanga,b,1, Rahul Mannana,b,1, Sethuramasundaram Pitchiayaa,b, Yuping Zhanga,b, Jesse W. Wotringd, Lanbo Xiaoa,b, Dan R. Robinsona,b, Yi-Mi Wua,b, Jean Ching-Yi Tiena,b, Xuhong Caoa,b,e, Stephanie A. Simkoa,b, Ingrid J. Apela,b, Pushpinder Bawaa,b, Steven Kregela,b, Sathiya P. Narayanana, Gregory Raskinda, Stephanie J. Ellisona, Abhijit Paroliaa,b, Sylvia Zelenka-Wanga,b, Lisa McMurrya,b, Fengyun Sua, Rui Wanga, Yunhui Chenga, Andrew D. Delektaa, Zejie Meif, Carla D. Prettog, Shaomeng Wanga,c,d,g,h, Rohit Mehraa,b,c,2, Jonathan Z. Sextond,g,i,j,2, and Arul M. Chinnaiyana,b,c,e,k,2,3 aMichigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI 48109; bDepartment of Pathology, University of Michigan, Ann Arbor, MI 48109; cRogel Cancer Center, University of Michigan, Ann Arbor, MI 48109; dDepartment of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109; eHoward Hughes Medical Institute, University of Michigan, Ann Arbor, MI 48109; fState Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Shanghai 200031, China; gDepartment of Internal Medicine, University of Michigan, Ann Arbor, MI 48109; hDepartment of Pharmacology, University of Michigan, Ann Arbor, MI 48109; iCenter for Drug Repurposing, University of Michigan, Ann Arbor, MI 48109; jMichigan Institute for Clinical and Health Research, University of Michigan, Ann Arbor, MI 48109; and kDepartment of Urology, University of Michigan, Ann Arbor, MI 48109 Contributed by Arul M.
    [Show full text]
  • Response and Resistance to BET Bromodomain Inhibitors in Triple Negative Breast Cancer
    HHS Public Access Author manuscript Author ManuscriptAuthor Manuscript Author Nature. Manuscript Author Author manuscript; Manuscript Author available in PMC 2016 July 06. Published in final edited form as: Nature. 2016 January 21; 529(7586): 413–417. doi:10.1038/nature16508. Response and resistance to BET bromodomain inhibitors in triple negative breast cancer Shaokun Shu#1,2, Charles Y. Lin#1,2, Housheng Hansen He#1,2,3,4,5, Robert M. Witwicki#1,2, Doris P. Tabassum1, Justin M. Roberts1, Michalina Janiszewska1,2, Sung Jin Huh1,2, Yi Liang4, Jeremy Ryan1,2, Ernest Doherty1,6, Hisham Mohammed7, Hao Guo3, Daniel G. Stover1,2, Muhammad B. Ekram1,2, Jonathan Brown1,2, Clive D'Santos7, Ian E. Krop1,2, Deborah Dillon1,8, Michael McKeown1,2, Christopher Ott1,2, Jun Qi1,2, Min Ni1,2, Prakash K. Rao9, Melissa Duarte9, Shwu-Yuan Wu10, Cheng-Ming Chiang10, Lars Anders11, Richard A. Young11, Eric Winer1,2, Antony Letai1,2, William T. Barry2,3, Jason S. Carroll7, Henry Long9, Myles Brown1,2,9, X. Shirley Liu3,9,12, Clifford A. Meyer1,2,3, James E. Bradner1,2,12, and Kornelia Polyak1,2,9,12 1Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA. 2Department of Medicine, Brigham and Women's Hospital, and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA. 3Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, and Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts USA. 4Princess Margaret Cancer Center/University Health Network, Toronto, Ontario, M5G1L7, Canada. 5Department of Medical Biophysics, University of Toronto, Toronto, Ontario, M5G2M9, Canada.
    [Show full text]
  • TG-1601 Combo a Demonstrate Correlated 2 Did Drug Mg/Kg, 94 (Blue Mg/Kg, a Anti 20 Mg/Kg Group Week
    TG-1601 is a novel BET inhibitor with strong binding affinity and long-lasting effect in preclinical models 5790 Emmanuel Normant1, Leonid Gorelik2, Rama Shmeis1, Henry Le1, Robert Nisch1, Karen TenHuisen1, Teja Turpuseema1, James Oliviero2, Dhanalakshmi Sivanandhan3, Payal Kiran Parikh3, Hari P. Miskin1, Peter Sportelli1 and Michael S. Weiss1 1) TG Therapeutics, New York, NY; 2) Checkpoint Therapeutics, New York, NY, 3) Jubilant Biosys, Bangalore, India Background In vitro and in vivo Pharmacodynamic activity In vivo anti-tumor activity Pharmacodynamic markers ❖ BET (bromodomain and extra-terminal) proteins bind to acetylated Kd (nM) In vitro pharmacodynamic activity of TG-1601 In vivo anti-tumor activity in MM1s multiple myeloma model In vivo and ex-vivo validation of CCR2 and IL1RN lysine residues on chromatin and participate in the regulation of gene TG-1601 and two related BET inhibitors bromodomain TG-1601 JQ1 OTX-015 transcription. Inhibition of BET protein binding to chromatin with small TG-1601 (µM) – 24h BRD2(BD1) 8.2 57 20 Left panel: Tumor volume; molecules selectively suppresses the transcription of a set of The black solid bar on the X oncogenes, including MYC and BCL-2. BRD2(BD2) 0.65 35 3 axis indicated the period of BRD3(BD1) 4 32 12 treatment. Right panel: Body weight changes compare to ❖ TG-1601 (aka CK-103) is a novel, selective and potent small molecule BRD3(BD2) 0.46 38 2 pre-dose weight. inhibitor of BET bromodomains. TG-1601 binds to the first and second BRD4(BD1) 11 31 13 The blue rectangles represent bromodomains (BD1, BD2) of the BET protein family, BRD2, BRD3, the interruption of dosing BRD4(BD2) 0.81 29 4.9 from Day 18 to Day 25 to BRD4, and BRDT, with Kd values ranging from 0.5 nM to 9.1 nM.
    [Show full text]
  • BET Protein Inhibitor JQ1 Downregulates Chromatin
    Zhou et al. Oncogenesis (2020) 9:33 https://doi.org/10.1038/s41389-020-0218-z Oncogenesis ARTICLE Open Access BET protein inhibitor JQ1 downregulates chromatin accessibility and suppresses metastasis of gastric cancer via inactivating RUNX2/NID1 signaling Siqi Zhou1,2, Shu Zhang2,3,LeiWang2,3, Shuling Huang2,3,YueYuan1,2,JieYang1,2,HuiWang2,3, Xihan Li2,3,PinWang2,3, Lin Zhou2,3,JunYang4,YuemeiXu4,HuanGao5, Yixuan Zhang2,3,YingLv2,3 and Xiaoping Zou1,2 Abstract Chromatin accessibility is critical for tumor development, whose mechanisms remain unclear. As a crucial regulator for chromatin remodeling, BRD4 promotes tumor progression by regulating multiple genes. As a small-molecule inhibitor of BRD4, JQ1 has potent chemotherapeutic activity against various human cancers. However, whether JQ1 has potential anti-tumor effects and how JQ1 regulates global transcription in gastric cancer (GC) remain largely unknown. In this research, we found BRD4 was highly expressed in GC tissues and was significantly associated with poor prognosis. JQ1 inhibited the proliferation and induced apoptosis of GC cells in vitro. Besides, JQ1 suppressed the migration and invasion of cancer cells by inducing MET. Notably, an assay for transposase-accessible chromatin using sequencing (ATAC-seq) data showed that JQ1 obviously downregulated the chromatin accessibility of GC cells and differentially accessible regions were highly enriched for RUNX2-binding motifs. Combinational analysis of ATAC-seq and RNA-seq 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; data discovered NID1 as the downstream target of JQ1 and JQ1 reduced NID1 expression in GC cells. Chromatin immunoprecipitation, luciferase reporter gene assay, and rescue experiments all confirmed that RUNX2/NID1 axis was responsible for JQ1-inhibiting metastasis of GC cells.
    [Show full text]
  • Macrophage Inflammatory Responses BET
    BET Protein Function Is Required for Inflammation: Brd2 Genetic Disruption and BET Inhibitor JQ1 Impair Mouse Macrophage Inflammatory Responses This information is current as of October 2, 2021. Anna C. Belkina, Barbara S. Nikolajczyk and Gerald V. Denis J Immunol 2013; 190:3670-3678; Prepublished online 18 February 2013; doi: 10.4049/jimmunol.1202838 Downloaded from http://www.jimmunol.org/content/190/7/3670 Supplementary http://www.jimmunol.org/content/suppl/2013/02/19/jimmunol.120283 Material 8.DC1 http://www.jimmunol.org/ References This article cites 51 articles, 19 of which you can access for free at: http://www.jimmunol.org/content/190/7/3670.full#ref-list-1 Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision by guest on October 2, 2021 • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2013 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology BET Protein Function Is Required for Inflammation: Brd2 Genetic Disruption and BET Inhibitor JQ1 Impair Mouse Macrophage Inflammatory Responses Anna C.
    [Show full text]
  • Loss of FBXW7-Mediated Degradation of BRAF Elicits Resistance to BET
    Yeh et al. Molecular Cancer (2020) 19:139 https://doi.org/10.1186/s12943-020-01254-x RESEARCH Open Access Loss of FBXW7-mediated degradation of BRAF elicits resistance to BET inhibitors in adult T cell leukemia cells Chien-Hung Yeh1, Marcia Bellon1, Fang Wang2, Hong Zhang2, Liwu Fu2 and Christophe Nicot1* Abstract Background: Human T cell leukemia virus type 1 (HTLV-1)-associated adult T cell leukemia (ATL) has a very poor prognosis with a median survival of 8 months and a 4-year overall survival of 11% for acute ATL. Present treatment options are limited and there is no curative therapy for ATL. Ubiquitin ligase FBXW7 is commonly mutated or functionally inactivated in human cancers. Consistent with the notion that FBXW7 controls the degradation of many oncoproteins, loss of FBXW7 has been linked to increased drug resistance or sensitivity in cancer cells. Method: In this study, we have characterized FBXW7 mutants previously identified in HTLV-I-infected ATL patient samples. TET-inducible ATL cells carrying wild type or mutated FBXW7 were analyzed for target degradation and for drug sensitivity. Results: Our results demonstrate that mutations in FBXW7 can selectively disrupt ubiquitination and proteasome degradation of target proteins: c-MYC, cyclin E and MCL1. Both c-MYC and MYCN were highly expressed in uncultured ATL patient’s samples and ATL-derived cell lines; and ATL cells demonstrated sensitivity to BET inhibitors in vitro and in vivo. High-throughput reverse phase protein array revealed BRAF as a novel target of FBXW7 and further experiments showed that mutations in FBXW7 preventing degradation of BRAF provided resistance to BET inhibitors.
    [Show full text]
  • BRD4 Is a Target for Treating Epileptic Seizures RU 1257 Technology Summary
    BRD4 is a target for treating epileptic seizures RU 1257 Technology Summary BRD4 is a member of the the Bromodomain and ExtraTerminal (BET) domain family of proteins which are responsible for “reading” acetylated lysine residues and regulating transcription in a variety of cell types in response to external signals. Aberrant BRD4/BET activity has been linked to a certain types of cancers which resulted in the development of a number of small molecule inhibitors, such as Jq1, that are currently being tested in clinical trials. However, very little is known about BRD4 activity in the brain. Studies carried out by Rockefeller University investigators Drs. Erica Korb, C. David Allis, and Robert B. Darnell showed that Jq1 caused changes in normal mouse neuronal cells by decreasing BRD4 activity, linking it to transcriptional processes important for learning and memory formation. To investigate whether BRD4 plays a role in neurological disorders, Dr. Korb and colleagues tested the Jq1 in a mouse model of chemically induced seizures – a condition caused by over-activity of the brain. The mice treated with the BRD4 inhibitor had a lower rate of seizures and mortality than mice that had not been treated with the drug. Ongoing research indicates that Brd4 inhibition also may be beneficial in other neurodevelopmental disorders that stem from similar causes. Overall these findings show significant promise for developing compounds specifically targeting BRD4 to help further probe the role of BRD4 in neurological diseases, and to develop novel therapeutics. Advantage Targeting BRD4 in neurodegenerative diseases is novel and underexplored Area of Application: Seizure susceptibility and memory formation Stage of Development: In vivo animal studies Lead Inventor: Dr.
    [Show full text]
  • The Bromodomain Protein BRD4 Positively Regulates Necroptosis Via Modulating MLKL Expression
    Cell Death & Differentiation (2019) 26:1929–1941 https://doi.org/10.1038/s41418-018-0262-9 ARTICLE The bromodomain protein BRD4 positively regulates necroptosis via modulating MLKL expression 1 2 1 2 1 1 1 1 Yu Xiong ● Linli Li ● Liting Zhang ● Yangyang Cui ● Chengyong Wu ● Hui Li ● Kai Chen ● Qiuyuan Yang ● 3 1 4 1 1 Rong Xiang ● Yiguo Hu ● Shile Huang ● Yuquan Wei ● Shengyong Yang Received: 8 July 2018 / Revised: 5 December 2018 / Accepted: 12 December 2018 / Published online: 15 January 2019 © ADMC Associazione Differenziamento e Morte Cellulare 2019 Abstract Necroptosis is a programmed form of necrotic cell death, which is tightly regulated by the necroptotic signaling pathway containing receptor-interacting protein (RIP)1, RIP3, and mixed-lineage kinase domain-like (MLKL) protein. In addition to the RIP1-RIP3-MLKL axis, other factors regulating necroptosis are still largely unknown. Here a cell-based small-molecule screening led to the finding that BET inhibitors protected cells from necroptosis in the TNFα/Smac-mimetic/Z-VAD-FMK (TSZ)-induced cell necroptosis model. Mechanistic studies revealed that BET inhibitors acted by downregulating MLKL expression. Further research demonstrated that BRD4, IRF1, P-TEFb, and RNA polymerase II formed a transcription 1234567890();,: 1234567890();,: complex to regulate the expression of MLKL, and BET inhibitors interfered with the transcription complex formation. In necroptosis-related disease model, the BET inhibitor JQ-1 showed promising therapeutic effects. Collectively, our studies establish, for the first time, BRD4 as a new epigenetic factor regulating necroptosis, and highlight the potential of BET inhibitors in the treatment of necroptosis-related diseases.
    [Show full text]