Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
PLoS One ; 17(3): e0262378, 2022.
Article in English | MEDLINE | ID: mdl-35353838

ABSTRACT

Therapeutic targeting of the estrogen receptor (ER) is a clinically validated approach for estrogen receptor positive breast cancer (ER+ BC), but sustained response is limited by acquired resistance. Targeting the transcriptional coactivators required for estrogen receptor activity represents an alternative approach that is not subject to the same limitations as targeting estrogen receptor itself. In this report we demonstrate that the acetyltransferase activity of coactivator paralogs CREBBP/EP300 represents a promising therapeutic target in ER+ BC. Using the potent and selective inhibitor CPI-1612, we show that CREBBP/EP300 acetyltransferase inhibition potently suppresses in vitro and in vivo growth of breast cancer cell line models and acts in a manner orthogonal to directly targeting ER. CREBBP/EP300 acetyltransferase inhibition suppresses ER-dependent transcription by targeting lineage-specific enhancers defined by the pioneer transcription factor FOXA1. These results validate CREBBP/EP300 acetyltransferase activity as a viable target for clinical development in ER+ breast cancer.


Subject(s)
Breast Neoplasms , Receptors, Estrogen , Acetyltransferases , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Cell Line, Tumor , Cell Proliferation , E1A-Associated p300 Protein/genetics , Female , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , MCF-7 Cells , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism
2.
Nat Chem Biol ; 18(2): 124-133, 2022 02.
Article in English | MEDLINE | ID: mdl-34952934

ABSTRACT

More than a decade after the launch of DNA methyltransferase and histone deacetylase inhibitors for the treatment of cancer, 2020 heralded the approval of the first histone methyltransferase inhibitor, revitalizing the concept that targeted manipulation of the chromatin regulatory landscape can have profound therapeutic impact. Three chromatin regulatory pathways-DNA methylation, histone acetylation and methylation-are frequently implicated in human cancer but hundreds of potentially druggable mechanisms complicate identification of key targets for therapeutic intervention. In addition to human genetics and functional screening, chemical biology approaches have proven critical for the discovery of key nodes in these pathways and in an ever-increasing complexity of molecularly defined human cancer contexts. This review introduces small molecule targeting approaches, showcases chemical probes and drug candidates for epigenetic writer enzymes, illustrates molecular features that may represent epigenetic dependencies and suggests translational strategies to maximize their impact in cancer therapy.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Chromatin/metabolism , Drug Delivery Systems , Gene Expression Regulation, Neoplastic/drug effects , Neoplasms/metabolism , Humans , Neoplasms/genetics
3.
Nat Rev Drug Discov ; 18(8): 609-628, 2019 08.
Article in English | MEDLINE | ID: mdl-31273347

ABSTRACT

Less than a decade ago, it was shown that bromodomains, acetyl lysine 'reader' modules found in proteins with varied functions, were highly tractable small-molecule targets. This is an unusual property for protein-protein or protein-peptide interaction domains, and it prompted a wave of chemical probe discovery to understand the biological potential of new agents that targeted bromodomains. The original examples, inhibitors of the bromodomain and extra-terminal (BET) class of bromodomains, showed enticing anti-inflammatory and anticancer activities, and several compounds have since advanced to human clinical trials. Here, we review the current state of BET inhibitor biology in relation to clinical development, and we discuss the next wave of bromodomain inhibitors with clinical potential in oncology and non-oncology indications. The lessons learned from BET inhibitor programmes should affect efforts to develop drugs that target non-BET bromodomains and other epigenetic readers.


Subject(s)
Drug Development/methods , Transcription Factors/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Clinical Trials as Topic , Epigenesis, Genetic , Humans , Molecular Targeted Therapy , Transcription Factors/genetics
4.
J Med Chem ; 59(23): 10549-10563, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27682507

ABSTRACT

The single bromodomain of the closely related transcriptional regulators CBP/EP300 is a target of much recent interest in cancer and immune system regulation. A co-crystal structure of a ligand-efficient screening hit and the CBP bromodomain guided initial design targeting the LPF shelf, ZA loop, and acetylated lysine binding regions. Structure-activity relationship studies allowed us to identify a more potent analogue. Optimization of permeability and microsomal stability and subsequent improvement of mouse hepatocyte stability afforded 59 (GNE-272, TR-FRET IC50 = 0.02 µM, BRET IC50 = 0.41 µM, BRD4(1) IC50 = 13 µM) that retained the best balance of cell potency, selectivity, and in vivo PK. Compound 59 showed a marked antiproliferative effect in hematologic cancer cell lines and modulates MYC expression in vivo that corresponds with antitumor activity in an AML tumor model.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Pyrazoles/pharmacology , Pyridones/pharmacology , p300-CBP Transcription Factors/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dogs , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Madin Darby Canine Kidney Cells , Mice , Mice, Nude , Models, Molecular , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyridones/chemical synthesis , Pyridones/chemistry , Structure-Activity Relationship
6.
Clin Cancer Res ; 22(16): 4259-70, 2016 Aug 15.
Article in English | MEDLINE | ID: mdl-27169995

ABSTRACT

PURPOSE: The initiation, progression, and maintenance of pancreatic ductal adenocarcinoma (PDAC) results from the interplay of genetic and epigenetic events. While the genetic alterations of PDAC have been well characterized, epigenetic pathways regulating PDAC remain, for the most part, elusive. The goal of this study was to identify novel epigenetic regulators contributing to the biology of PDAC. EXPERIMENTAL DESIGN: In vivo pooled shRNA screens targeting 118 epigenetic proteins were performed in two orthotopic PDAC xenograft models. Candidate genes were characterized in 19 human PDAC cell lines, heterotopic xenograft tumor models, and a genetically engineered mouse (GEM) model of PDAC. Gene expression, IHC, and immunoprecipitation experiments were performed to analyze the pathways by which candidate genes contribute to PDAC. RESULTS: In vivo shRNA screens identified BRD2 and BRD3, members of the BET family of chromatin adaptors, as key regulators of PDAC tumor growth. Pharmacologic inhibition of BET bromodomains enhanced survival in a PDAC GEM model and inhibited growth of human-derived xenograft tumors. BET proteins contribute to PDAC cell growth through direct interaction with members of the GLI family of transcription factors and modulating their activity. Within cancer cells, BET bromodomain inhibition results in downregulation of SHH, a key mediator of the tumor microenvironment and canonical activator of GLI. Consistent with this, inhibition of BET bromodomains decreases cancer-associated fibroblast content of tumors in both GEM and xenograft tumor models. CONCLUSIONS: Therapeutic inhibition of BET proteins offers a novel mechanism to target both the neoplastic and stromal components of PDAC. Clin Cancer Res; 22(16); 4259-70. ©2016 AACR.


Subject(s)
Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , RNA-Binding Proteins/metabolism , Tumor Microenvironment , Zinc Finger Protein GLI1/metabolism , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Gene Expression Profiling , Genes, myc , Hedgehog Proteins/metabolism , Heterografts , Humans , Mice , Pancreatic Neoplasms/genetics , Protein Binding , Protein Interaction Domains and Motifs , RNA Interference , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/chemistry , Signal Transduction , Tumor Burden , Tumor Microenvironment/genetics
7.
ACS Med Chem Lett ; 7(5): 531-6, 2016 May 12.
Article in English | MEDLINE | ID: mdl-27190605

ABSTRACT

CBP and EP300 are highly homologous, bromodomain-containing transcription coactivators involved in numerous cellular pathways relevant to oncology. As part of our effort to explore the potential therapeutic implications of selectively targeting bromodomains, we set out to identify a CBP/EP300 bromodomain inhibitor that was potent both in vitro and in cellular target engagement assays and was selective over the other members of the bromodomain family. Reported here is a series of cell-potent and selective probes of the CBP/EP300 bromodomains, derived from the fragment screening hit 4-methyl-1,3,4,5-tetrahydro-2H-benzo[b][1,4]diazepin-2-one.

8.
J Biol Chem ; 291(25): 13014-27, 2016 Jun 17.
Article in English | MEDLINE | ID: mdl-27056325

ABSTRACT

Covalent modification of histones is a fundamental mechanism of regulated gene expression in eukaryotes, and interpretation of histone modifications is an essential feature of epigenetic control. Bromodomains are specialized binding modules that interact with acetylated histones, linking chromatin recognition to gene transcription. Because of their ability to function in a domain-specific fashion, selective disruption of bromodomain:acetylated histone interactions with chemical probes serves as a powerful means for understanding biological processes regulated by these chromatin adaptors. Here we describe the discovery and characterization of potent and selective small molecule inhibitors for the bromodomains of CREBBP/EP300 that engage their target in cellular assays. We use these tools to demonstrate a critical role for CREBBP/EP300 bromodomains in regulatory T cell biology. Because regulatory T cell recruitment to tumors is a major mechanism of immune evasion by cancer cells, our data highlight the importance of CREBBP/EP300 bromodomain inhibition as a novel, small molecule-based approach for cancer immunotherapy.


Subject(s)
CREB-Binding Protein/antagonists & inhibitors , E1A-Associated p300 Protein/antagonists & inhibitors , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , T-Lymphocytes, Regulatory/drug effects , Acetylation/drug effects , CREB-Binding Protein/chemistry , CREB-Binding Protein/metabolism , Cell Differentiation/drug effects , Cell Line , Cells, Cultured , E1A-Associated p300 Protein/chemistry , E1A-Associated p300 Protein/metabolism , Forkhead Transcription Factors/metabolism , Histones/metabolism , Humans , Molecular Docking Simulation , Protein Structure, Tertiary/drug effects , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Transcriptome/drug effects
9.
Elife ; 52016 Jan 05.
Article in English | MEDLINE | ID: mdl-26731516

ABSTRACT

Pharmacological inhibition of chromatin co-regulatory factors represents a clinically validated strategy to modulate oncogenic signaling through selective attenuation of gene expression. Here, we demonstrate that CBP/EP300 bromodomain inhibition preferentially abrogates the viability of multiple myeloma cell lines. Selective targeting of multiple myeloma cell lines through CBP/EP300 bromodomain inhibition is the result of direct transcriptional suppression of the lymphocyte-specific transcription factor IRF4, which is essential for the viability of myeloma cells, and the concomitant repression of the IRF4 target gene c-MYC. Ectopic expression of either IRF4 or MYC antagonizes the phenotypic and transcriptional effects of CBP/EP300 bromodomain inhibition, highlighting the IRF4/MYC axis as a key component of its mechanism of action. These findings suggest that CBP/EP300 bromodomain inhibition represents a viable therapeutic strategy for targeting multiple myeloma and other lymphoid malignancies dependent on the IRF4 network.


Subject(s)
Antineoplastic Agents/pharmacology , E1A-Associated p300 Protein/antagonists & inhibitors , Interferon Regulatory Factors/metabolism , Multiple Myeloma/physiopathology , Peptide Fragments/antagonists & inhibitors , Sialoglycoproteins/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Humans
10.
Cancer Res ; 76(6): 1313-9, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26759243

ABSTRACT

Small-molecule inhibitors of the bromodomain and extraterminal (BET) family of proteins are being tested in clinical trials for a variety of cancers, but patient selection strategies remain limited. This challenge is partly attributed to the heterogeneous responses elicited by BET inhibition (BETi), including cellular differentiation, senescence, and death. In this study, we performed phenotypic and gene-expression analyses of treatment-naive and engineered tolerant cell lines representing human melanoma and leukemia to elucidate the dominant features defining response to BETi. We found that de novo and acquired tolerance to BETi is driven by the robustness of the apoptotic response, and that genetic or pharmacologic manipulation of the apoptotic signaling network can modify the phenotypic response to BETi. We further reveal that the expression signatures of the apoptotic genes BCL2, BCL2L1, and BAD significantly predict response to BETi. Taken together, our findings highlight the apoptotic program as a determinant of response to BETi, and provide a molecular basis for patient stratification and combination therapy development.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Small Molecule Libraries/pharmacology , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HL-60 Cells , HT29 Cells , Humans , K562 Cells , Leukemia/drug therapy , Leukemia/metabolism , Melanoma/drug therapy , Melanoma/metabolism , Signal Transduction/drug effects
11.
Proc Natl Acad Sci U S A ; 108(40): 16669-74, 2011 Oct 04.
Article in English | MEDLINE | ID: mdl-21949397

ABSTRACT

The MYC transcription factor is a master regulator of diverse cellular functions and has been long considered a compelling therapeutic target because of its role in a range of human malignancies. However, pharmacologic inhibition of MYC function has proven challenging because of both the diverse mechanisms driving its aberrant expression and the challenge of disrupting protein-DNA interactions. Here, we demonstrate the rapid and potent abrogation of MYC gene transcription by representative small molecule inhibitors of the BET family of chromatin adaptors. MYC transcriptional suppression was observed in the context of the natural, chromosomally translocated, and amplified gene locus. Inhibition of BET bromodomain-promoter interactions and subsequent reduction of MYC transcript and protein levels resulted in G(1) arrest and extensive apoptosis in a variety of leukemia and lymphoma cell lines. Exogenous expression of MYC from an artificial promoter that is resistant to BET regulation significantly protected cells from cell cycle arrest and growth suppression by BET inhibitors. MYC suppression was accompanied by deregulation of the MYC transcriptome, including potent reactivation of the p21 tumor suppressor. Treatment with a BET inhibitor resulted in significant antitumor activity in xenograft models of Burkitt's lymphoma and acute myeloid leukemia. These findings demonstrate that pharmacologic inhibition of MYC is achievable through targeting BET bromodomains. Such inhibitors may have clinical utility given the widespread pathogenetic role of MYC in cancer.


Subject(s)
Apoptosis/physiology , Burkitt Lymphoma/drug therapy , Cell Cycle/physiology , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/physiology , Leukemia, Myeloid, Acute/drug therapy , Transcription Factors/metabolism , Animals , Apoptosis/genetics , Azepines/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Polymerase Chain Reaction , Protein Structure, Tertiary/genetics , RNA, Small Interfering/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Triazoles/pharmacology
12.
Genes Dev ; 25(8): 814-30, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21498571

ABSTRACT

pRB-mediated inhibition of cell proliferation is a complex process that depends on the action of many proteins. However, little is known about the specific pathways that cooperate with the Retinoblastoma protein (pRB) and the variables that influence pRB's ability to arrest tumor cells. Here we describe two shRNA screens that identify kinases that are important for pRB to suppress cell proliferation and pRB-mediated induction of senescence markers. The results reveal an unexpected effect of LATS2, a component of the Hippo pathway, on pRB-induced phenotypes. Partial knockdown of LATS2 strongly suppresses some pRB-induced senescence markers. Further analysis shows that LATS2 cooperates with pRB to promote the silencing of E2F target genes, and that reduced levels of LATS2 lead to defects in the assembly of DREAM (DP, RB [retinoblastoma], E2F, and MuvB) repressor complexes at E2F-regulated promoters. Kinase assays show that LATS2 can phosphorylate DYRK1A, and that it enhances the ability of DYRK1A to phosphorylate the DREAM subunit LIN52. Intriguingly, the LATS2 locus is physically linked with RB1 on 13q, and this region frequently displays loss of heterozygosity in human cancers. Our results reveal a functional connection between the pRB and Hippo tumor suppressor pathways, and suggest that low levels of LATS2 may undermine the ability of pRB to induce a permanent cell cycle arrest in tumor cells.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/physiology , Retinoblastoma Protein/metabolism , Tumor Suppressor Proteins/metabolism , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line , Cell Line, Tumor , Cell Proliferation , Chromatin Immunoprecipitation , Flow Cytometry , Humans , Immunoblotting , Loss of Heterozygosity/genetics , Oligonucleotide Array Sequence Analysis , Phosphorylation , Polymerase Chain Reaction , Protein Binding/genetics , Protein Binding/physiology , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA, Small Interfering/genetics , Retinoblastoma Protein/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Tumor Suppressor Proteins/genetics , Dyrk Kinases
13.
Proc Natl Acad Sci U S A ; 107(35): 15455-60, 2010 Aug 31.
Article in English | MEDLINE | ID: mdl-20713694

ABSTRACT

Chromosomal instability and the subsequent genetic mutations are considered to be critical factors in the development of the majority of solid tumors, but the mechanisms by which a stable diploid cell loses the ability to maintain genomic integrity are not well characterized. We have approached this critical issue through the use of high-throughput screens in untransformed diploid epithelial cells. In a screen of a cDNA library, we identified 13 kinases whose overexpression leads to increased ploidy. In a series of shRNA screens, we identified 16 kinases whose loss leads to increased ploidy. In both cDNA and shRNA screens, the majority of hits have not been linked previously to genomic stability. We further show that sustained loss of the shRNA screening hits leads to multipolar spindles and heterogeneous chromosome content, two characteristics of chromosomal instability. Loss of several of the kinases leads to loss of contact inhibition and to anchorage-independent growth, vital traits acquired during tumor development. We anticipate that this work will serve as a template for the comprehensive identification of pathways whose dysregulation can drive tumorigenesis through impaired karyotypic maintenance.


Subject(s)
Chromosomal Instability/genetics , Diploidy , Epithelial Cells/metabolism , Protein Kinases/genetics , Aneuploidy , Cell Line , Epithelial Cells/cytology , Gene Expression Profiling/methods , Gene Library , Humans , In Situ Hybridization, Fluorescence , Mitosis/genetics , Models, Genetic , RNA Interference
14.
Proc Natl Acad Sci U S A ; 107(35): 15461-6, 2010 Aug 31.
Article in English | MEDLINE | ID: mdl-20713695

ABSTRACT

Chromosomal instability and the subsequent genetic mutations are considered to be critical factors in the development of the majority of solid tumors. Here, we describe how the nucleoside diphosphate kinase Nm23-H1, a protein with a known link to cancer progression, regulates a critical step during cytokinesis. Nm23-H1 acts to provide a local source of GTP for the GTPase dynamin. Loss of Nm23-H1 in diploid cells leads to cytokinetic furrow regression, followed by cytokinesis failure and generation of tetraploid cells. Loss of dynamin phenocopies loss of Nm23-H1, and ectopic overexpression of WT dynamin complements the loss of Nm23-H1. In the absence of p53 signaling, the tetraploid cells resulting from loss of Nm23-H1 continue cycling and develop classic hallmarks of tumor cells. We thus provide evidence that the loss of Nm23-H1, an event suspected to promote metastasis, may additionally function at an earlier stage of tumor development to drive the acquisition of chromosomal instability.


Subject(s)
Chromosomal Instability/genetics , Cytokinesis/genetics , Dynamins/genetics , NM23 Nucleoside Diphosphate Kinases/genetics , Blotting, Western , Cell Cycle/genetics , Cell Line , Cells, Cultured , Cellular Senescence/genetics , Dynamins/metabolism , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Microscopy, Fluorescence , Mitosis/genetics , NM23 Nucleoside Diphosphate Kinases/metabolism , Polyploidy , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
15.
Nat Cell Biol ; 6(4): 366-72, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15104092

ABSTRACT

Transforming growth factor beta (TGF-beta) induces both apoptosis and cell-cycle arrest in some cell lines, but only growth arrest in others. It is not clear how this differential response to TGF-beta is specified. Smad proteins are critical mediators of TGF-beta signalling. After stimulation by TGF-beta, Smad2 and Smad3 become phosphorylated by the activated TGF-beta receptor kinases, oligomerize with Smad4, translocate to the nucleus and regulate the expression of TGF-beta target genes. Here we report that the sensitivity to TGF-beta induced apoptosis is regulated by crosstalk between the Akt/PKB serine/threonine kinase and Smad3 through a mechanism that is independent of Akt kinase activity. Akt interacts directly with unphosphorylated Smad3 to sequester it outside the nucleus, preventing its phosphorylation and nuclear translocation. This results in inhibition of Smad3-mediated transcription and apoptosis. Furthermore, the ratio of Smad3 to Akt correlates with the sensitivity of cells to TGF-beta induced apoptosis. Alteration of this ratio changes the apoptotic, but not the growth-inhibitory, responses of cells to TGF-beta. These findings identify an important determinant of sensitivity to TGF-beta-induced apoptosis that involves crosstalk between the TGF-beta and phosphatidylinositol-3-OH kinase (PI(3)K) pathways.


Subject(s)
Apoptosis/genetics , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Signal Transduction/genetics , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Apoptosis/drug effects , Cell Line , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , DNA-Binding Proteins/genetics , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Signal Transduction/drug effects , Smad3 Protein , Trans-Activators/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...