Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Cells ; 12(13)2023 06 22.
Article in English | MEDLINE | ID: mdl-37443729

ABSTRACT

The SMYD family is a unique class of lysine methyltransferases (KMTases) whose catalytic SET domain is split by a MYND domain. Among these, Smyd1 was identified as a heart- and skeletal muscle-specific KMTase and is essential for cardiogenesis and skeletal muscle development. SMYD1 has been characterized as a histone methyltransferase (HMTase). Here we demonstrated that SMYD1 methylates is the Skeletal muscle-specific splice variant of the Nascent polypeptide-Associated Complex (skNAC) transcription factor. SMYD1-mediated methylation of skNAC targets K1975 within the carboxy-terminus region of skNAC. Catalysis requires physical interaction of SMYD1 and skNAC via the conserved MYND domain of SMYD1 and the PXLXP motif of skNAC. Our data indicated that skNAC methylation is required for the direct transcriptional activation of myoglobin (Mb), a heart- and skeletal muscle-specific hemoprotein that facilitates oxygen transport. Our study revealed that the skNAC, as a methylation target of SMYD1, illuminates the molecular mechanism by which SMYD1 cooperates with skNAC to regulate transcriptional activation of genes crucial for muscle functions and implicates the MYND domain of the SMYD-family KMTases as an adaptor to target substrates for methylation.


Subject(s)
DNA-Binding Proteins , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase , Molecular Chaperones , Muscle Development , Muscle Proteins , Transcription Factors , Transcriptional Activation , Humans , Catalysis , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HEK293 Cells , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Methylation , Molecular Chaperones/metabolism , Muscle Development/genetics , Muscle Proteins/chemistry , Muscle Proteins/genetics , Muscle Proteins/metabolism , Mutation , Protein Domains , Protein Isoforms/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism
2.
J Med Chem ; 65(16): 11177-11186, 2022 08 25.
Article in English | MEDLINE | ID: mdl-35930799

ABSTRACT

Bromodomains are acetyllysine recognition domains present in a variety of human proteins. Bromodomains also bind small molecules that compete with acetyllysine, and therefore bromodomains have been targets for drug discovery efforts. Highly potent and selective ligands with good cellular permeability have been proposed as chemical probes for use in exploring the functions of many of the bromodomain proteins. We report here the discovery of a class of such inhibitors targeting the family VIII bromodomains of SMARCA2 (BRM) and SMARCA4 (BRG1), and PBRM1 (polybromo-1) bromodomain 5. We propose one example from this series, GNE-064, as a chemical probe for the bromodomains SMARCA2, SMARCA4, and PBRM1(5) with the potential for in vivo use.


Subject(s)
DNA Helicases , Transcription Factors , DNA-Binding Proteins , Humans , Nuclear Proteins , Protein Domains
3.
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
4.
J Biol Chem ; 296: 100349, 2021.
Article in English | MEDLINE | ID: mdl-33524394

ABSTRACT

The histone methyltransferase EZH2 has been the target of numerous small-molecule inhibitor discovery efforts over the last 10+ years. Emerging clinical data have provided early evidence for single agent activity with acceptable safety profiles for first-generation inhibitors. We have developed kinetic methodologies for studying EZH2-inhibitor-binding kinetics that have allowed us to identify a unique structural modification that results in significant increases in the drug-target residence times of all EZH2 inhibitor scaffolds we have studied. The unexpected residence time enhancement bestowed by this modification has enabled us to create a series of second-generation EZH2 inhibitors with sub-pM binding affinities. We provide both biophysical evidence validating this sub-pM potency and biological evidence demonstrating the utility and relevance of such high-affinity interactions with EZH2.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Allosteric Regulation/drug effects , Animals , Drug Discovery , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , HeLa Cells , Humans , Mice, SCID , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
5.
ACS Med Chem Lett ; 11(6): 1205-1212, 2020 Jun 11.
Article in English | MEDLINE | ID: mdl-32551002

ABSTRACT

Histone methyltransferase EZH2, which is the catalytic subunit of the PRC2 complex, catalyzes the methylation of histone H3K27-a transcriptionally repressive post-translational modification (PTM). EZH2 is commonly mutated in hematologic malignancies and frequently overexpressed in solid tumors, where its expression level often correlates with poor prognosis. First generation EZH2 inhibitors are beginning to show clinical benefit, and we believe that a second generation EZH2 inhibitor could further build upon this foundation to fully realize the therapeutic potential of EZH2 inhibition. During our medicinal chemistry campaign, we identified 4-thiomethyl pyridone as a key modification that led to significantly increased potency and prolonged residence time. Leveraging this finding, we optimized a series of EZH2 inhibitors, with enhanced antitumor activity and improved physiochemical properties, which have the potential to expand the clinical use of EZH2 inhibition.

6.
ACS Med Chem Lett ; 11(6): 1324-1329, 2020 Jun 11.
Article in English | MEDLINE | ID: mdl-32551019

ABSTRACT

The histone acetyltransferases, CREB binding protein (CBP) and EP300, are master transcriptional co-regulators that have been implicated in numerous diseases, such as cancer, inflammatory disorders, and neurodegeneration. A novel, highly potent, orally bioavailable EP300/CBP histone acetyltransferase (HAT) inhibitor, CPI-1612 or 17, was developed from the lead compound 3. Replacement of the indole scaffold of 3 with the aminopyridine scaffold of 17 led to improvements in potency, solubility, and bioavailability. These characteristics resulted in a 20-fold lower efficacious dose for 17 relative to lead 3 in a JEKO-1 tumor mouse xenograft study.

7.
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
8.
Fluids Barriers CNS ; 15(1): 22, 2018 Aug 16.
Article in English | MEDLINE | ID: mdl-30111340

ABSTRACT

BACKGROUND: Choroid plexus epithelial cells express high levels of transthyretin, produce cerebrospinal fluid and many of its proteins, and make up the blood-cerebrospinal fluid barrier. Choroid plexus epithelial cells are vital to brain health and may be involved in neurological diseases. Transgenic mice containing fluorescent and luminescent reporters of these cells would facilitate their study in health and disease, but prior transgenic reporters lost expression over the early postnatal period. METHODS: Human bacterial artificial chromosomes in which the transthyretin coding sequence was replaced with DNA for tdTomato or luciferase 2 were used in pronuclear injections to produce transgenic mice. These mice were characterized by visualizing red fluorescence, immunostaining, real-time reverse transcription polymerase chain reaction, and luciferase enzyme assay. RESULTS: Reporters were faithfully expressed in cells that express transthyretin constitutively, including choroid plexus epithelial cells, retinal pigment epithelium, pancreatic islets, and liver. Expression of tdTomato in choroid plexus began at the appropriate embryonic age, being detectable by E11.5. Relative levels of tdTomato transcript in the liver and choroid plexus paralleled relative levels of transcripts for transthyretin. Expression remained robust over the first postnatal year, although choroid plexus transcripts of tdTomato declined slightly with age whereas transthyretin remained constant. TdTomato expression patterns were consistent across three founder lines, displayed no sex differences, and were stable across several generations. Two of the tdTomato lines were bred to homozygosity, and homozygous mice are healthy and fertile. The usefulness of tdTomato reporters in visualizing and analyzing live Transwell cultures was demonstrated. Luciferase activity was very high in homogenates of choroid plexus and continued to be expressed through adulthood. Luciferase also was detectable in eye and pancreas. CONCLUSIONS: Transgenic mice bearing fluorescent and luminescent reporters of transthyretin should prove useful for tracking transplanted choroid plexus epithelial cells, for purifying the cells, and for reporting their derivation from stem cells. They also should prove useful for studying transthyretin synthesis by other cell types, as transthyretin has been implicated in many functions and conditions, including clearance of ß-amyloid peptides associated with Alzheimer's disease, heat shock in neurons, processing of neuropeptides, nerve regeneration, astrocyte metabolism, and transthyretin amyloidosis.


Subject(s)
Choroid Plexus/cytology , Epithelial Cells/cytology , Luminescent Proteins/metabolism , Mice, Transgenic , Models, Animal , Prealbumin/metabolism , Animals , Cell Culture Techniques , Cells, Cultured , Choroid Plexus/growth & development , Choroid Plexus/metabolism , Chromosomes, Artificial, Bacterial , Epithelial Cells/metabolism , Humans , Islets of Langerhans/cytology , Islets of Langerhans/growth & development , Islets of Langerhans/metabolism , Liver/cytology , Liver/growth & development , Liver/metabolism , Luminescent Proteins/genetics , Prealbumin/genetics , RNA, Messenger/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/growth & development , Retinal Pigment Epithelium/metabolism
9.
ACS Med Chem Lett ; 8(7): 737-741, 2017 Jul 13.
Article in English | MEDLINE | ID: mdl-28740608

ABSTRACT

The biological function of bromodomains, epigenetic readers of acetylated lysine residues, remains largely unknown. Herein we report our efforts to discover a potent and selective inhibitor of the bromodomain of cat eye syndrome chromosome region candidate 2 (CECR2). Screening of our internal medicinal chemistry collection led to the identification of a pyrrolopyridone chemical lead, and subsequent structure-based drug design led to a potent and selective CECR2 bromodomain inhibitor (GNE-886) suitable for use as an in vitro tool compound.

11.
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
12.
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.

13.
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
14.
ACS Med Chem Lett ; 7(2): 145-50, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26985289

ABSTRACT

Inhibition of the bromodomains of the BET family, of which BRD4 is a member, has been shown to decrease myc and interleukin (IL) 6 in vivo, markers that are of therapeutic relevance to cancer and inflammatory disease, respectively. Herein we report substituted benzo[b]isoxazolo[4,5-d]azepines and benzotriazolo[4,3-d][1,4]diazepines as fragment-derived novel inhibitors of the bromodomain of BRD4. Compounds from these series were potent and selective in cells, and subsequent optimization of microsomal stability yielded representatives that demonstrated dose- and time-dependent reduction of plasma IL-6 in mice.

15.
J Med Chem ; 59(4): 1330-9, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26815195

ABSTRACT

In recent years, inhibition of the interaction between the bromodomain and extra-terminal domain (BET) family of chromatin adaptors and acetyl-lysine residues on chromatin has emerged as a promising approach to regulate the expression of important disease-relevant genes, including MYC, BCL-2, and NF-κB. Here we describe the identification and characterization of a potent and selective benzoisoxazoloazepine BET bromodomain inhibitor that attenuates BET-dependent gene expression in vivo, demonstrates antitumor efficacy in an MV-4-11 mouse xenograft model, and is currently undergoing human clinical trials for hematological malignancies (CPI-0610).


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Azepines/chemistry , Azepines/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Leukemia, Myeloid, Acute/drug therapy , Nuclear Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Azepines/pharmacokinetics , Azepines/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , Clinical Trials as Topic , Dogs , Genes, myc/drug effects , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Rats , Transcription Factors/chemistry , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
16.
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
17.
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
18.
Mol Cell ; 58(2): 323-38, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25843623

ABSTRACT

Excess dormant origins bound by the minichromosome maintenance (MCM) replicative helicase complex play a critical role in preventing replication stress, chromosome instability, and tumorigenesis. In response to DNA damage, replicating cells must coordinate DNA repair and dormant origin firing to ensure complete and timely replication of the genome; how cells regulate this process remains elusive. Herein, we identify a member of the Fanconi anemia (FA) DNA repair pathway, FANCI, as a key effector of dormant origin firing in response to replication stress. Cells lacking FANCI have reduced number of origins, increased inter-origin distances, and slowed proliferation rates. Intriguingly, ATR-mediated FANCI phosphorylation inhibits dormant origin firing while promoting replication fork restart/DNA repair. Using super-resolution microscopy, we show that FANCI co-localizes with MCM-bound chromatin in response to replication stress. These data reveal a unique role for FANCI as a modulator of dormant origin firing and link timely genome replication to DNA repair.


Subject(s)
Chromatin/metabolism , DNA Damage , DNA Replication , Fanconi Anemia Complementation Group Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Cell Proliferation , Fanconi Anemia Complementation Group Proteins/genetics , HeLa Cells , Humans , Hydroxyurea/pharmacology , Minichromosome Maintenance Proteins/genetics , Minichromosome Maintenance Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
19.
Bioorg Med Chem Lett ; 25(9): 1842-8, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25851940

ABSTRACT

In this report we detail the evolution of our previously reported thiophene isoxazole BET inhibitor chemotype exemplified by CPI-3 to a novel bromodomain selective chemotype (the methyl isoxazoleazepine chemotype) exemplified by carboxamide 23. The methyl isoxazoleazepine chemotype provides potent inhibition of the bromodomains of the BET family, excellent in vivo PK across species, low unbound clearance, and target engagement in a MYC PK-PD model.


Subject(s)
Azepines/pharmacology , Drug Design , Nuclear Proteins/antagonists & inhibitors , Oxazoles/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Azepines/chemical synthesis , Azepines/chemistry , Cell Cycle Proteins , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Oxazoles/chemical synthesis , Oxazoles/chemistry , Structure-Activity Relationship
20.
ACS Med Chem Lett ; 4(9): 835-40, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24900758

ABSTRACT

The identification of a novel series of small molecule BET inhibitors is described. Using crystallographic binding modes of an amino-isoxazole fragment and known BET inhibitors, a structure-based drug design effort lead to a novel isoxazole azepine scaffold. This scaffold showed good potency in biochemical and cellular assays and oral activity in an in vivo model of BET inhibition.

SELECTION OF CITATIONS
SEARCH DETAIL
...