Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Med Chem ; 63(2): 714-746, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31904959

ABSTRACT

The bromodomain and extraterminal (BET) family of bromodomain-containing proteins are important regulators of the epigenome through their ability to recognize N-acetyl lysine (KAc) post-translational modifications on histone tails. These interactions have been implicated in various disease states and, consequently, disruption of BET-KAc binding has emerged as an attractive therapeutic strategy with a number of small molecule inhibitors now under investigation in the clinic. However, until the utility of these advanced candidates is fully assessed by these trials, there remains scope for the discovery of inhibitors from new chemotypes with alternative physicochemical, pharmacokinetic, and pharmacodynamic profiles. Herein, we describe the discovery of a candidate-quality dimethylpyridone benzimidazole compound which originated from the hybridization of a dimethylphenol benzimidazole series, identified using encoded library technology, with an N-methyl pyridone series identified through fragment screening. Optimization via structure- and property-based design led to I-BET469, which possesses favorable oral pharmacokinetic properties, displays activity in vivo, and is projected to have a low human efficacious dose.


Subject(s)
High-Throughput Screening Assays/methods , Proteins/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzimidazoles/chemistry , Benzimidazoles/pharmacokinetics , Benzimidazoles/pharmacology , Chemokine CCL2/biosynthesis , Crystallography, X-Ray , Drug Discovery , Drug Evaluation, Preclinical , Drug Synergism , Humans , Interleukin-6/antagonists & inhibitors , Leukocytes/drug effects , Male , Mice , Models, Molecular , Protein Processing, Post-Translational/drug effects , Small Molecule Libraries
2.
Science ; 356(6345): 1397-1401, 2017 06 30.
Article in English | MEDLINE | ID: mdl-28619718

ABSTRACT

The success of new therapies hinges on our ability to understand their molecular and cellular mechanisms of action. We modified BET bromodomain inhibitors, an epigenetic-based therapy, to create functionally conserved compounds that are amenable to click chemistry and can be used as molecular probes in vitro and in vivo. We used click proteomics and click sequencing to explore the gene regulatory function of BRD4 (bromodomain containing protein 4) and the transcriptional changes induced by BET inhibitors. In our studies of mouse models of acute leukemia, we used high-resolution microscopy and flow cytometry to highlight the heterogeneity of drug activity within tumor cells located in different tissue compartments. We also demonstrate the differential distribution and effects of BET inhibitors in normal and malignant cells in vivo. This study provides a potential framework for the preclinical assessment of a wide range of drugs.


Subject(s)
Benzodiazepines/therapeutic use , Click Chemistry , Drug Delivery Systems , Epigenomics , Leukemia/drug therapy , Animals , Benzodiazepines/pharmacology , Cells, Cultured , Disease Models, Animal , Leukemia/pathology , Mice , Precision Medicine , Tissue Distribution , Transcription Factors/antagonists & inhibitors
3.
Bioorg Med Chem Lett ; 26(19): 4606-4612, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27578246

ABSTRACT

The optimisation of the azanaphthyridine series of Spleen Tyrosine Kinase inhibitors is described. The medicinal chemistry strategy was focused on optimising the human whole blood activity whilst achieving a sufficient margin over hERG activity. A good pharmacokinetic profile was achieved by modification of the pKa. Morpholine compound 32 is a potent SYK inhibitor showing moderate selectivity, good oral bioavailability and good efficacy in the rat Arthus model but demonstrated a genotoxic potential in the Ames assay.


Subject(s)
Naphthyridines/pharmacology , Protein Kinase Inhibitors/pharmacology , Administration, Oral , Animals , Biological Availability , Crystallography, X-Ray , Humans , Mutagenicity Tests , Naphthyridines/administration & dosage , Naphthyridines/pharmacokinetics , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Rats , Structure-Activity Relationship
4.
Nat Struct Mol Biol ; 23(7): 673-81, 2016 07.
Article in English | MEDLINE | ID: mdl-27294782

ABSTRACT

Targeted therapies against disruptor of telomeric silencing 1-like (DOT1L) and bromodomain-containing protein 4 (BRD4) are currently being evaluated in clinical trials. However, the mechanisms by which BRD4 and DOT1L regulate leukemogenic transcription programs remain unclear. Using quantitative proteomics, chemoproteomics and biochemical fractionation, we found that native BRD4 and DOT1L exist in separate protein complexes. Genetic disruption or small-molecule inhibition of BRD4 and DOT1L showed marked synergistic activity against MLL leukemia cell lines, primary human leukemia cells and mouse leukemia models. Mechanistically, we found a previously unrecognized functional collaboration between DOT1L and BRD4 that is especially important at highly transcribed genes in proximity to superenhancers. DOT1L, via dimethylated histone H3 K79, facilitates histone H4 acetylation, which in turn regulates the binding of BRD4 to chromatin. These data provide new insights into the regulation of transcription and specify a molecular framework for therapeutic intervention in this disease with poor prognosis.


Subject(s)
Gene Expression Regulation, Leukemic , Histones/genetics , Leukemia, Biphenotypic, Acute/genetics , Methyltransferases/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Acetylation , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Cycle Proteins , Cell Proliferation , Chromatin/chemistry , Chromatin/metabolism , Clinical Trials as Topic , Disease Models, Animal , Female , Histone-Lysine N-Methyltransferase , Histones/metabolism , Humans , Leukemia, Biphenotypic, Acute/metabolism , Leukemia, Biphenotypic, Acute/pathology , Male , Methyltransferases/antagonists & inhibitors , Methyltransferases/metabolism , Mice , Mice, Inbred C57BL , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Primary Cell Culture , Protein Binding , Proteomics/methods , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Transcription, Genetic
5.
Cancer Res ; 75(23): 5106-5119, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26552700

ABSTRACT

The histone acetyltransferases CBP/p300 are involved in recurrent leukemia-associated chromosomal translocations and are key regulators of cell growth. Therefore, efforts to generate inhibitors of CBP/p300 are of clinical value. We developed a specific and potent acetyl-lysine competitive protein-protein interaction inhibitor, I-CBP112, that targets the CBP/p300 bromodomains. Exposure of human and mouse leukemic cell lines to I-CBP112 resulted in substantially impaired colony formation and induced cellular differentiation without significant cytotoxicity. I-CBP112 significantly reduced the leukemia-initiating potential of MLL-AF9(+) acute myeloid leukemia cells in a dose-dependent manner in vitro and in vivo. Interestingly, I-CBP112 increased the cytotoxic activity of BET bromodomain inhibitor JQ1 as well as doxorubicin. Collectively, we report the development and preclinical evaluation of a novel, potent inhibitor targeting CBP/p300 bromodomains that impairs aberrant self-renewal of leukemic cells. The synergistic effects of I-CBP112 and current standard therapy (doxorubicin) as well as emerging treatment strategies (BET inhibition) provide new opportunities for combinatorial treatment of leukemia and potentially other cancers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Enzyme Inhibitors/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Oxazepines/pharmacology , p300-CBP Transcription Factors/antagonists & inhibitors , Amino Acid Sequence , Animals , Cell Line, Tumor , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Synergism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Humans , Leukemia, Myeloid, Acute/enzymology , Mice , Models, Molecular , Molecular Sequence Data , Oxazepines/administration & dosage , Oxazepines/chemistry , Protein Structure, Tertiary , Xenograft Model Antitumor Assays , p300-CBP Transcription Factors/chemistry
6.
Nature ; 525(7570): 538-42, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26367796

ABSTRACT

Bromodomain and extra terminal protein (BET) inhibitors are first-in-class targeted therapies that deliver a new therapeutic opportunity by directly targeting bromodomain proteins that bind acetylated chromatin marks. Early clinical trials have shown promise, especially in acute myeloid leukaemia, and therefore the evaluation of resistance mechanisms is crucial to optimize the clinical efficacy of these drugs. Here we use primary mouse haematopoietic stem and progenitor cells immortalized with the fusion protein MLL-AF9 to generate several single-cell clones that demonstrate resistance, in vitro and in vivo, to the prototypical BET inhibitor, I-BET. Resistance to I-BET confers cross-resistance to chemically distinct BET inhibitors such as JQ1, as well as resistance to genetic knockdown of BET proteins. Resistance is not mediated through increased drug efflux or metabolism, but is shown to emerge from leukaemia stem cells both ex vivo and in vivo. Chromatin-bound BRD4 is globally reduced in resistant cells, whereas the expression of key target genes such as Myc remains unaltered, highlighting the existence of alternative mechanisms to regulate transcription. We demonstrate that resistance to BET inhibitors, in human and mouse leukaemia cells, is in part a consequence of increased Wnt/ß-catenin signalling, and negative regulation of this pathway results in restoration of sensitivity to I-BET in vitro and in vivo. Together, these findings provide new insights into the biology of acute myeloid leukaemia, highlight potential therapeutic limitations of BET inhibitors, and identify strategies that may enhance the clinical utility of these unique targeted therapies.


Subject(s)
Benzodiazepines/pharmacology , Drug Resistance, Neoplasm/drug effects , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Nuclear Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Animals , Azepines/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , Cells, Cultured , Chromatin/metabolism , Clone Cells/drug effects , Clone Cells/metabolism , Clone Cells/pathology , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/drug effects , Genes, myc/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Molecular Targeted Therapy , Neoplastic Stem Cells/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Triazoles/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
7.
PLoS One ; 8(11): e80746, 2013.
Article in English | MEDLINE | ID: mdl-24260471

ABSTRACT

Recent studies suggest that BET inhibitors are effective anti-cancer therapeutics. Here we show that BET inhibitors are effective against murine primary mammary tumors, but not pulmonary metastases. BRD4, a target of BET inhibitors, encodes two isoforms with opposite effects on tumor progression. To gain insights into why BET inhibition was ineffective against metastases the pro-metastatic short isoform of BRD4 was characterized using mass spectrometry and cellular fractionation. Our data show that the pro-metastatic short isoform interacts with the LINC complex and the metastasis-associated proteins RRP1B and SIPA1 at the inner face of the nuclear membrane. Furthermore, histone binding arrays revealed that the short isoform has a broader acetylated histone binding pattern relative to the long isoform. These differential biochemical and nuclear localization properties revealed in our study provide novel insights into the opposing roles of BRD4 isoforms in metastatic breast cancer progression.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , GTPase-Activating Proteins/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Cycle Proteins , Cell Line, Tumor , Cell Nucleus/metabolism , Disease Models, Animal , Female , Heterocyclic Compounds, 4 or More Rings/pharmacology , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Microtubule-Associated Proteins/metabolism , N-Terminal Acetyltransferase E/metabolism , N-Terminal Acetyltransferases , Neoplasm Metastasis , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/genetics , Protein Binding , Protein Isoforms , Protein Transport , Transcription Factors/genetics , Tumor Burden/drug effects
8.
Gut ; 62(11): 1581-90, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23242119

ABSTRACT

OBJECTIVE: Intestinal inflammation resulting from manipulation-induced mast cell activation is a crucial mechanism in the pathophysiology of postoperative ileus (POI). Recently it has been shown that spleen tyrosine kinase (Syk) is involved in mast cell degranulation. Therefore, we have evaluated the effect of the Syk-inhibitor GSK compound 143 (GSK143) as potential treatment to shorten POI. DESIGN: In vivo: in a mouse model of POI, the effect of the Syk inhibitor (GSK143) was evaluated on gastrointestinal transit, muscular inflammation and cytokine production. In vitro: the effect of GSK143 and doxantrazole were evaluated on cultured peritoneal mast cells (PMCs) and bone marrow derived macrophages. RESULTS: In vivo: intestinal manipulation resulted in a delay in gastrointestinal transit at t=24 h (Geometric Center (GC): 4.4 ± 0.3). Doxantrazole and GSK143 significantly increased gastrointestinal transit (GC doxantrazole (10 mg/kg): 7.2 ± 0.7; GSK143 (1 mg/kg): 7.6 ± 0.6), reduced inflammation and prevented recruitment of immune cells in the intestinal muscularis. In vitro: in PMCs, substance P (0-90 µM) and trinitrophenyl (0-4 µg/ml) induced a concentration-dependent release of ß-hexosaminidase. Pretreatment with doxantrazole and GSK143 (0.03-10 µM) concentration dependently blocked substance P and trinitrophenyl induced ß-hexosaminidase release. In addition, GSK143 was able to reduce cytokine expression in endotoxin-treated bone marrow derived macrophages in a concentration-dependent manner. CONCLUSIONS: The Syk inhibitor GSK143 reduces macrophage activation and mast cell degranulation in vitro. In addition, it inhibits manipulation-induced intestinal muscular inflammation and restores intestinal transit in mice. These findings suggest that Syk inhibition may be a new tool to shorten POI.


Subject(s)
Aniline Compounds/therapeutic use , Ileus/prevention & control , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Postoperative Complications/prevention & control , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/therapeutic use , Aniline Compounds/administration & dosage , Aniline Compounds/pharmacology , Animals , Cell Degranulation/drug effects , Cells, Cultured , Cytokines/biosynthesis , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Gastrointestinal Transit/drug effects , Ileus/physiopathology , Macrophage Activation/drug effects , Mast Cells/drug effects , Mast Cells/physiology , Mice , Mice, Inbred C57BL , Ovalbumin/antagonists & inhibitors , Ovalbumin/pharmacology , Phosphodiesterase Inhibitors/therapeutic use , Postoperative Complications/physiopathology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Substance P/antagonists & inhibitors , Substance P/pharmacology , Syk Kinase , Thioxanthenes/therapeutic use , Xanthones/therapeutic use
9.
Nature ; 478(7370): 529-33, 2011 Oct 02.
Article in English | MEDLINE | ID: mdl-21964340

ABSTRACT

Recurrent chromosomal translocations involving the mixed lineage leukaemia (MLL) gene initiate aggressive forms of leukaemia, which are often refractory to conventional therapies. Many MLL-fusion partners are members of the super elongation complex (SEC), a critical regulator of transcriptional elongation, suggesting that aberrant control of this process has an important role in leukaemia induction. Here we use a global proteomic strategy to demonstrate that MLL fusions, as part of SEC and the polymerase-associated factor complex (PAFc), are associated with the BET family of acetyl-lysine recognizing, chromatin 'adaptor' proteins. These data provided the basis for therapeutic intervention in MLL-fusion leukaemia, via the displacement of the BET family of proteins from chromatin. We show that a novel small molecule inhibitor of the BET family, GSK1210151A (I-BET151), has profound efficacy against human and murine MLL-fusion leukaemic cell lines, through the induction of early cell cycle arrest and apoptosis. I-BET151 treatment in two human leukaemia cell lines with different MLL fusions alters the expression of a common set of genes whose function may account for these phenotypic changes. The mode of action of I-BET151 is, at least in part, due to the inhibition of transcription at key genes (BCL2, C-MYC and CDK6) through the displacement of BRD3/4, PAFc and SEC components from chromatin. In vivo studies indicate that I-BET151 has significant therapeutic value, providing survival benefit in two distinct mouse models of murine MLL-AF9 and human MLL-AF4 leukaemia. Finally, the efficacy of I-BET151 against human leukaemia stem cells is demonstrated, providing further evidence of its potent therapeutic potential. These findings establish the displacement of BET proteins from chromatin as a promising epigenetic therapy for these aggressive leukaemias.


Subject(s)
Chromatin/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Chromatin/genetics , Chromatin Immunoprecipitation , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Binding/drug effects , Proteomics , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...