Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Med Chem ; 65(2): 1458-1480, 2022 01 27.
Article in English | MEDLINE | ID: mdl-34726887

ABSTRACT

CDK7 has emerged as an exciting target in oncology due to its roles in two important processes that are misregulated in cancer cells: cell cycle and transcription. This report describes the discovery of SY-5609, a highly potent (sub-nM CDK7 Kd) and selective, orally available inhibitor of CDK7 that entered the clinic in 2020 (ClinicalTrials.gov Identifier: NCT04247126). Structure-based design was leveraged to obtain high selectivity (>4000-times the closest off target) and slow off-rate binding kinetics desirable for potent cellular activity. Finally, incorporation of a phosphine oxide as an atypical hydrogen bond acceptor helped provide the required potency and metabolic stability. The development candidate SY-5609 displays potent inhibition of CDK7 in cells and demonstrates strong efficacy in mouse xenograft models when dosed as low as 2 mg/kg.


Subject(s)
Breast Neoplasms , Cell Cycle , Cyclin-Dependent Kinases , Drug Discovery , Protein Kinase Inhibitors , Animals , Female , Humans , Mice , Apoptosis , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Cyclin-Dependent Kinase-Activating Kinase , Cyclin-Dependent Kinases/antagonists & inhibitors , Mice, Inbred BALB C , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Genes Dev ; 34(21-22): 1452-1473, 2020 11 01.
Article in English | MEDLINE | ID: mdl-33060135

ABSTRACT

CDK7 associates with the 10-subunit TFIIH complex and regulates transcription by phosphorylating the C-terminal domain (CTD) of RNA polymerase II (RNAPII). Few additional CDK7 substrates are known. Here, using the covalent inhibitor SY-351 and quantitative phosphoproteomics, we identified CDK7 kinase substrates in human cells. Among hundreds of high-confidence targets, the vast majority are unique to CDK7 (i.e., distinct from other transcription-associated kinases), with a subset that suggest novel cellular functions. Transcription-associated factors were predominant CDK7 substrates, including SF3B1, U2AF2, and other splicing components. Accordingly, widespread and diverse splicing defects, such as alternative exon inclusion and intron retention, were characterized in CDK7-inhibited cells. Combined with biochemical assays, we establish that CDK7 directly activates other transcription-associated kinases CDK9, CDK12, and CDK13, invoking a "master regulator" role in transcription. We further demonstrate that TFIIH restricts CDK7 kinase function to the RNAPII CTD, whereas other substrates (e.g., SPT5 and SF3B1) are phosphorylated by the three-subunit CDK-activating kinase (CAK; CCNH, MAT1, and CDK7). These results suggest new models for CDK7 function in transcription and implicate CAK dissociation from TFIIH as essential for kinase activation. This straightforward regulatory strategy ensures CDK7 activation is spatially and temporally linked to transcription, and may apply toward other transcription-associated kinases.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Models, Biological , Transcription Factor TFIIH/metabolism , Transcription, Genetic/genetics , Alternative Splicing/genetics , Cell Survival/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Enzyme Activation/genetics , HL-60 Cells , Humans , Cyclin-Dependent Kinase-Activating Kinase
3.
Cancer Res ; 79(13): 3479-3491, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31064851

ABSTRACT

Recent studies suggest that targeting transcriptional machinery can lead to potent and selective anticancer effects in cancers dependent on high and constant expression of certain transcription factors for growth and survival. Cyclin-dependent kinase 7 (CDK7) is the catalytic subunit of the CDK-activating kinase complex. Its function is required for both cell-cycle regulation and transcriptional control of gene expression. CDK7 has recently emerged as an attractive cancer target because its inhibition leads to decreased transcript levels of oncogenic transcription factors, especially those associated with super-enhancers. Here, we describe a selective CDK7 inhibitor SY-1365, which is currently in clinical trials in populations of patients with ovarian and breast cancer (NCT03134638). In vitro, SY-1365 inhibited cell growth of many different cancer types at nanomolar concentrations. SY-1365 treatment decreased MCL1 protein levels, and cancer cells with low BCL2L1 (BCL-XL) expression were found to be more sensitive to SY-1365. Transcriptional changes in acute myeloid leukemia (AML) cell lines were distinct from those following treatment with other transcriptional inhibitors. SY-1365 demonstrated substantial antitumor effects in multiple AML xenograft models as a single agent; SY-1365-induced growth inhibition was enhanced in combination with the BCL2 inhibitor venetoclax. Antitumor activity was also observed in xenograft models of ovarian cancer, suggesting the potential for exploring SY-1365 in the clinic in both hematologic and solid tumors. Our findings support targeting CDK7 as a new approach for treating transcriptionally addicted cancers. SIGNIFICANCE: These findings demonstrate the molecular mechanism of action and potent antitumor activity of SY-1365, the first selective CDK7 inhibitor to enter clinical investigation.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Ovarian Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Animals , Cell Cycle/drug effects , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Female , High-Throughput Screening Assays , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Models, Molecular , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/enzymology , Protein Kinase Inhibitors/chemistry , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Cyclin-Dependent Kinase-Activating Kinase
4.
Blood ; 128(14): 1845-1853, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27531676

ABSTRACT

The oncogenic transcription factor signal transducer and activator of transcription 3 (STAT3) is frequently activated inappropriately in a wide range of hematological and solid cancers, but clinically available therapies targeting STAT3 are lacking. Using a computational strategy to identify compounds opposing the gene expression signature of STAT3, we discovered atovaquone (Mepron), an antimicrobial approved by the US Food and Drug Administration, to be a potent STAT3 inhibitor. We show that, at drug concentrations routinely achieved clinically in human plasma, atovaquone inhibits STAT3 phosphorylation, the expression of STAT3 target genes, and the viability of STAT3-dependent hematological cancer cells. These effects were also observed with atovaquone treatment of primary blasts isolated from patients with acute myelogenous leukemia or acute lymphocytic leukemia. Atovaquone is not a kinase inhibitor but instead rapidly and specifically downregulates cell-surface expression of glycoprotein 130, which is required for STAT3 activation in multiple contexts. The administration of oral atovaquone to mice inhibited tumor growth and prolonged survival in a murine model of multiple myeloma. Finally, in patients with acute myelogenous leukemia treated with hematopoietic stem cell transplantation, extended use of atovaquone for Pneumocystis prophylaxis was associated with improved relapse-free survival. These findings establish atovaquone as a novel, clinically accessible STAT3 inhibitor with evidence of anticancer efficacy in both animal models and humans.


Subject(s)
Antineoplastic Agents/pharmacology , Atovaquone/pharmacology , Drug Discovery , Gene Expression Regulation, Neoplastic/drug effects , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Atovaquone/chemistry , Atovaquone/therapeutic use , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Cytokine Receptor gp130/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Phosphorylation/drug effects , Phosphotyrosine/metabolism , STAT3 Transcription Factor/metabolism , Treatment Outcome
5.
Anal Chem ; 87(12): 6349-56, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26023704

ABSTRACT

Analysis of complex mixtures of proteins by hydrogen exchange (HX) mass spectrometry (MS) is limited by one's ability to resolve the protein(s) of interest from the proteins that are not of interest. One strategy for overcoming this problem is to tag the target protein(s) to allow for rapid removal from the mixture for subsequent analysis. Here we illustrate a new solution involving fluorous conjugation of a retrievable probe. The appended fluorous tag allows for facile immobilization on a fluorous surface. When a target protein is passed over the immobilized probe molecule, it can be efficiently captured and then exposed to a flowing stream of deuterated buffer for hydrogen exchange. The utility of this method is illustrated for a model system of the Elongin BC protein complex bound to a peptide from HIV Vif. Efficient capture is demonstrated, and deuteration when immobilized was identical to deuteration in conventional solution-phase hydrogen exchange MS. Protein captured from a crude bacterial cell lysate could also be deuterated without the need for separate purification steps before HX MS. The advantages and disadvantages of the method are discussed in light of miniaturization and automation.


Subject(s)
Fluorocarbons/chemistry , Hydrogen/chemistry , Molecular Probe Techniques , Proteins/analysis , Hydrocarbons, Fluorinated , Mass Spectrometry , Solutions
6.
ACS Chem Biol ; 10(3): 667-74, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25397901

ABSTRACT

The DOT1L lysine methyltransferase has emerged as a validated therapeutic target in MLL-rearranged (MLLr) acute leukemias. Although S-adenosylmethionine competitive inhibitors have demonstrated pharmacological proof-of-principle in MLLr-leukemia, these compounds require further optimization to improve cellular potency and pharmacokinetic stability. Limiting DOT1L inhibitor discovery and ligand optimization have been complex biochemical methods often using radionucleotides and cellular methods requiring prolonged culture. We therefore developed a new suite of assay technologies that allows comparative assessment of chemical tools for DOT1L in a miniaturized format. Coupling these assays with structural information, we developed new insights into DOT1L ligand binding and identified several functionalized probes with increased cellular potency (IC50 values ∼10 nM) and excellent selectivity for DOT1L. Together these assay technologies define a platform capability for discovery and optimization of small-molecule DOT1L inhibitors.


Subject(s)
Antineoplastic Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Gene Expression Regulation, Neoplastic , High-Throughput Screening Assays , Histones/antagonists & inhibitors , Methyltransferases/antagonists & inhibitors , Adenosine/analogs & derivatives , Adenosine/chemical synthesis , Adenosine/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelial Cells/pathology , Histone-Lysine N-Methyltransferase , Histones/genetics , Histones/metabolism , Humans , Ligands , Methylation , Methyltransferases/chemistry , Methyltransferases/genetics , Methyltransferases/metabolism , Models, Molecular , Phenylurea Compounds/chemical synthesis , Phenylurea Compounds/pharmacology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , S-Adenosylmethionine/chemistry , S-Adenosylmethionine/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
7.
J Med Chem ; 57(21): 9019-27, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25314271

ABSTRACT

BET bromodomain inhibition has contributed new insights into gene regulation and emerged as a promising therapeutic strategy in cancer. Structural analogy of early methyl-triazolo BET inhibitors has prompted a need for structurally dissimilar ligands as probes of bromodomain function. Using fluorous-tagged multicomponent reactions, we developed a focused chemical library of bromodomain inhibitors around a 3,5-dimethylisoxazole biasing element with micromolar biochemical IC50. Iterative synthesis and biochemical assessment allowed optimization of novel BET bromodomain inhibitors based on an imidazo[1,2-a]pyrazine scaffold. Lead compound 32 (UMB-32) binds BRD4 with a Kd of 550 nM and 724 nM cellular potency in BRD4-dependent lines. Additionally, compound 32 shows potency against TAF1, a bromodomain-containing transcription factor previously unapproached by discovery chemistry. Compound 32 was cocrystallized with BRD4, yielding a 1.56 Å resolution crystal structure. This research showcases new applications of fluorous and multicomponent chemical synthesis for the development of novel epigenetic inhibitors.


Subject(s)
Imidazoles/chemical synthesis , Isoxazoles/chemistry , Nuclear Proteins/antagonists & inhibitors , Pyrazines/chemical synthesis , Transcription Factors/antagonists & inhibitors , Alkanesulfonic Acids/chemistry , Azepines/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , Crystallography, X-Ray , Fluorocarbons/chemistry , Gene Expression Regulation , Humans , Imidazoles/pharmacology , Inhibitory Concentration 50 , Ligands , Models, Molecular , Nuclear Proteins/chemistry , Pyrazines/pharmacology , Pyridines/chemical synthesis , Small Molecule Libraries/chemical synthesis , Structure-Activity Relationship , Transcription Factors/chemistry
8.
Bioorg Med Chem Lett ; 24(16): 4035-8, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25011911

ABSTRACT

7,9-Diaryl-1,6,8-trioxaspiro[4.5]dec-3-en-2-ones are a recently described group of spirocyclic butenolides that can be generated rapidly and as a single diastereomer through a cascade process between γ-hydroxybutenolides and aromatic aldehydes. The following outlines our findings that these spirocycles are potently cytotoxic and have a dramatic structure-function profile that provides excellent insight into the structural features required for this potency.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Spiro Compounds/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Bacillus subtilis/drug effects , Bacillus subtilis/growth & development , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Escherichia coli/drug effects , Escherichia coli/growth & development , Humans , Microbial Sensitivity Tests , Molecular Conformation , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/growth & development , Spiro Compounds/chemical synthesis , Spiro Compounds/chemistry , Structure-Activity Relationship
9.
J Clin Invest ; 124(2): 644-55, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24401270

ABSTRACT

T cell acute lymphoblastic leukemia (T-ALL) is an aggressive cancer that is frequently associated with activating mutations in NOTCH1 and dysregulation of MYC. Here, we performed 2 complementary screens to identify FDA-approved drugs and drug-like small molecules with activity against T-ALL. We developed a zebrafish system to screen small molecules for toxic activity toward MYC-overexpressing thymocytes and used a human T-ALL cell line to screen for small molecules that synergize with Notch inhibitors. We identified the antipsychotic drug perphenazine in both screens due to its ability to induce apoptosis in fish, mouse, and human T-ALL cells. Using ligand-affinity chromatography coupled with mass spectrometry, we identified protein phosphatase 2A (PP2A) as a perphenazine target. T-ALL cell lines treated with perphenazine exhibited rapid dephosphorylation of multiple PP2A substrates and subsequent apoptosis. Moreover, shRNA knockdown of specific PP2A subunits attenuated perphenazine activity, indicating that PP2A mediates the drug's antileukemic activity. Finally, human T-ALLs treated with perphenazine exhibited suppressed cell growth and dephosphorylation of PP2A targets in vitro and in vivo. Our findings provide a mechanistic explanation for the recurring identification of phenothiazines as a class of drugs with anticancer effects. Furthermore, these data suggest that pharmacologic PP2A activation in T-ALL and other cancers driven by hyperphosphorylated PP2A substrates has therapeutic potential.


Subject(s)
Apoptosis , Phenothiazines/chemistry , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Phosphatase 2/metabolism , Animals , Animals, Genetically Modified , Cell Line, Tumor , Cell Survival , Chromatography, Affinity , Disease Models, Animal , Dopamine Antagonists/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Mass Spectrometry , Mice , Perphenazine/chemistry , Phosphorylation , Pigmentation , Proteomics , Receptors, Notch/metabolism , Time Factors , Zebrafish
10.
Nat Biotechnol ; 32(1): 92-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24336317

ABSTRACT

A vast number of small-molecule ligands, including therapeutic drugs under development and in clinical use, elicit their effects by binding specific proteins associated with the genome. An ability to map the direct interactions of a chemical entity with chromatin genome-wide could provide important insights into chemical perturbation of cellular function. Here we describe a method that couples ligand-affinity capture and massively parallel DNA sequencing (Chem-seq) to identify the sites bound by small chemical molecules throughout the human genome. We show how Chem-seq can be combined with ChIP-seq to gain unique insights into the interaction of drugs with their target proteins throughout the genome of tumor cells. These methods will be broadly useful to enhance understanding of therapeutic action and to characterize the specificity of chemical entities that interact with DNA or genome-associated proteins.


Subject(s)
Chromatin/genetics , DNA/genetics , Proteins/genetics , Transcription Factors/genetics , Binding Sites/genetics , Genome, Human , High-Throughput Nucleotide Sequencing , Humans , Ligands , Protein Binding/genetics
11.
Nat Commun ; 3: 1288, 2012.
Article in English | MEDLINE | ID: mdl-23250418

ABSTRACT

Selective inhibition of protein methyltransferases is a promising new approach to drug discovery. An attractive strategy towards this goal is the development of compounds that selectively inhibit binding of the cofactor, S-adenosylmethionine, within specific protein methyltransferases. Here we report the three-dimensional structure of the protein methyltransferase DOT1L bound to EPZ004777, the first S-adenosylmethionine-competitive inhibitor of a protein methyltransferase with in vivo efficacy. This structure and those of four new analogues reveal remodelling of the catalytic site. EPZ004777 and a brominated analogue, SGC0946, inhibit DOT1L in vitro and selectively kill mixed lineage leukaemia cells, in which DOT1L is aberrantly localized via interaction with an oncogenic MLL fusion protein. These data provide important new insight into mechanisms of cell-active S-adenosylmethionine-competitive protein methyltransferase inhibitors, and establish a foundation for the further development of drug-like inhibitors of DOT1L for cancer therapy.


Subject(s)
Methyltransferases/antagonists & inhibitors , Adenosine/analogs & derivatives , Adenosine/pharmacology , Binding, Competitive/drug effects , Blotting, Western , Catalysis , Catalytic Domain/drug effects , Histone-Lysine N-Methyltransferase , Humans , Kinetics , Methyltransferases/metabolism , Phenylurea Compounds/pharmacology , Structure-Activity Relationship , Substrate Specificity , Surface Plasmon Resonance
12.
ACS Comb Sci ; 14(6): 343-6, 2012 Jun 11.
Article in English | MEDLINE | ID: mdl-22578077

ABSTRACT

Cycloadditions of cyclobutadiene can offer rapid access to rigid polycyclic ring systems. Further functionalization of these strained-ring cycloadducts can lead to unique scaffolds for probing unexplored regions of chemical space. Along these lines, opportunities for high-throughput syntheses of these novel systems could be facilitated with the introduction of an immobilized cyclobutadiene reagent. Reported herein are preliminary studies of an iron tricarbonyl cyclobutadiene complex attached to solid support. Oxidative unmasking of the immobilized cyclobutadiene in the presence of various dienophiles is shown to produce a small collection of substituted bicyclo[2.2.0]hexene derivatives. The solid support cycloaddition strategy is shown to be comparable, but lower in efficiency to solution phase methods for generating these cycloadducts.


Subject(s)
Alkadienes/chemistry , Alkenes/chemistry , Bridged Bicyclo Compounds/chemical synthesis , Butanes/chemistry , Solid-Phase Synthesis Techniques/methods , Alkenes/chemical synthesis , Bridged Bicyclo Compounds/chemistry , Cycloaddition Reaction , Iron Compounds/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...