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










Publication year range
1.
Cancer Cell ; 36(1): 100-114.e25, 2019 07 08.
Article in English | MEDLINE | ID: mdl-31257072

ABSTRACT

Type I protein arginine methyltransferases (PRMTs) catalyze asymmetric dimethylation of arginines on proteins. Type I PRMTs and their substrates have been implicated in human cancers, suggesting inhibition of type I PRMTs may offer a therapeutic approach for oncology. The current report describes GSK3368715 (EPZ019997), a potent, reversible type I PRMT inhibitor with anti-tumor effects in human cancer models. Inhibition of PRMT5, the predominant type II PRMT, produces synergistic cancer cell growth inhibition when combined with GSK3368715. Interestingly, deletion of the methylthioadenosine phosphorylase gene (MTAP) results in accumulation of the metabolite 2-methylthioadenosine, an endogenous inhibitor of PRMT5, and correlates with sensitivity to GSK3368715 in cell lines. These data provide rationale to explore MTAP status as a biomarker strategy for patient selection.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Purine-Nucleoside Phosphorylase/deficiency , Alternative Splicing , Antineoplastic Agents/chemistry , Biomarkers , Cell Line, Tumor , Drug Synergism , Enzyme Inhibitors/chemistry , Humans , Methylation , Models, Molecular , Molecular Conformation , Molecular Structure , Protein Binding , Protein-Arginine N-Methyltransferases/chemistry , Substrate Specificity
2.
Sci Rep ; 7(1): 17993, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29269946

ABSTRACT

CARM1 is an arginine methyltransferase with diverse histone and non-histone substrates implicated in the regulation of cellular processes including transcriptional co-activation and RNA processing. CARM1 overexpression has been reported in multiple cancer types and has been shown to modulate oncogenic pathways in in vitro studies. Detailed understanding of the mechanism of action of CARM1 in oncogenesis has been limited by a lack of selective tool compounds, particularly for in vivo studies. We describe the identification and characterization of, to our knowledge, the first potent and selective inhibitor of CARM1 that exhibits anti-proliferative effects both in vitro and in vivo and, to our knowledge, the first demonstration of a role for CARM1 in multiple myeloma (MM). EZM2302 (GSK3359088) is an inhibitor of CARM1 enzymatic activity in biochemical assays (IC50 = 6 nM) with broad selectivity against other histone methyltransferases. Treatment of MM cell lines with EZM2302 leads to inhibition of PABP1 and SMB methylation and cell stasis with IC50 values in the nanomolar range. Oral dosing of EZM2302 demonstrates dose-dependent in vivo CARM1 inhibition and anti-tumor activity in an MM xenograft model. EZM2302 is a validated chemical probe suitable for further understanding the biological role CARM1 plays in cancer and other diseases.


Subject(s)
Antineoplastic Agents/therapeutic use , CARD Signaling Adaptor Proteins/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Guanylate Cyclase/antagonists & inhibitors , Isoxazoles/therapeutic use , Multiple Myeloma/drug therapy , Pyrimidines/therapeutic use , Spiro Compounds/therapeutic use , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacokinetics , Humans , In Vitro Techniques , Isoxazoles/pharmacokinetics , Male , Mice , Neoplasm Transplantation , Pyrimidines/pharmacokinetics , Rats, Sprague-Dawley , Spiro Compounds/pharmacokinetics
3.
Cell Signal ; 40: 73-80, 2017 12.
Article in English | MEDLINE | ID: mdl-28867658

ABSTRACT

We previously constructed a collection of fission yeast strains that express various mammalian cyclic nucleotide phosphodiesterases (PDEs) and developed a cell-based high throughput screen (HTS) for small molecule PDE inhibitors. Here we describe a compound, BC54, that is a selective inhibitor of enzymes from the cAMP-specific PDE4 and PDE7 families. Consistent with the biological effect of other PDE4 and PDE7 inhibitors, BC54 displays potent anti-inflammatory properties and is superior to a combination of rolipram (a PDE4 inhibitor) and BRL50481 (a PDE7A inhibitor) for inducing apoptosis in chronic lymphocytic leukemia (CLL) cells. We further exploited PKA-regulated growth phenotypes in fission yeast to isolate two mutant alleles of the human PDE4B2 gene that encode enzymes possessing single amino acid changes that confer partial resistance to BC54. We confirm this resistance to both BC54 and rolipram via yeast-based assays and, for PDE4B2T407A, in vitro enzyme assays. Thus, we are able to use this system for both chemical screens to identify biologically-active PDE inhibitors and molecular genetic studies to characterize the interaction of these molecules with their target enzymes. Based on its potency, selectivity, and effectiveness in cell culture, BC54 should be a useful tool to study biological processes regulated by PDE4 and PDE7 enzymes.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 7/antagonists & inhibitors , Cyclohexanes/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Apoptosis/drug effects , Cyclic AMP/metabolism , Drug Evaluation, Preclinical , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Rolipram/administration & dosage , Schizosaccharomyces/drug effects , Schizosaccharomyces/genetics
4.
J Med Chem ; 59(4): 1556-64, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26769278

ABSTRACT

Posttranslational methylation of histones plays a critical role in gene regulation. Misregulation of histone methylation can lead to oncogenic transformation. Enhancer of Zeste homologue 2 (EZH2) methylates histone 3 at lysine 27 (H3K27) and abnormal methylation of this site is found in many cancers. Tazemetostat, an EHZ2 inhibitor in clinical development, has shown activity in both preclinical models of cancer as well as in patients with lymphoma or INI1-deficient solid tumors. Herein we report the structure-activity relationships from identification of an initial hit in a high-throughput screen through selection of tazemetostat for clinical development. The importance of several methyl groups to the potency of the inhibitors is highlighted as well as the importance of balancing pharmacokinetic properties with potency.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Histones/metabolism , Methylation/drug effects , Polycomb Repressive Complex 2/antagonists & inhibitors , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Animals , Drug Discovery , Enhancer of Zeste Homolog 2 Protein , Enzyme Inhibitors/pharmacokinetics , Humans , Mice , Polycomb Repressive Complex 2/metabolism , Protein Processing, Post-Translational/drug effects , Small Molecule Libraries/pharmacokinetics , Structure-Activity Relationship
5.
Mol Cancer Ther ; 13(4): 842-54, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24563539

ABSTRACT

Mutations within the catalytic domain of the histone methyltransferase EZH2 have been identified in subsets of patients with non-Hodgkin lymphoma (NHL). These genetic alterations are hypothesized to confer an oncogenic dependency on EZH2 enzymatic activity in these cancers. We have previously reported the discovery of EPZ005678 and EPZ-6438, potent and selective S-adenosyl-methionine-competitive small molecule inhibitors of EZH2. Although both compounds are similar with respect to their mechanism of action and selectivity, EPZ-6438 possesses superior potency and drug-like properties, including good oral bioavailability in animals. Here, we characterize the activity of EPZ-6438 in preclinical models of NHL. EPZ-6438 selectively inhibits intracellular lysine 27 of histone H3 (H3K27) methylation in a concentration- and time-dependent manner in both EZH2 wild-type and mutant lymphoma cells. Inhibition of H3K27 trimethylation (H3K27Me3) leads to selective cell killing of human lymphoma cell lines bearing EZH2 catalytic domain point mutations. Treatment of EZH2-mutant NHL xenograft-bearing mice with EPZ-6438 causes dose-dependent tumor growth inhibition, including complete and sustained tumor regressions with correlative diminution of H3K27Me3 levels in tumors and selected normal tissues. Mice dosed orally with EPZ-6438 for 28 days remained tumor free for up to 63 days after stopping compound treatment in two EZH2-mutant xenograft models. These data confirm the dependency of EZH2-mutant NHL on EZH2 activity and portend the utility of EPZ-6438 as a potential treatment for these genetically defined cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Lymphoma, Non-Hodgkin/drug therapy , Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb Repressive Complex 2/genetics , Pyridones/pharmacology , Animals , Apoptosis/drug effects , Biphenyl Compounds , Catalytic Domain/genetics , Cell Cycle/drug effects , Cell Line, Tumor , Female , Humans , Lymphoma, Non-Hodgkin/pathology , Male , Mice , Mice, SCID , Molecular Sequence Data , Morpholines , Point Mutation , Rats , Rats, Sprague-Dawley , Xenograft Model Antitumor Assays
6.
Biopharm Drug Dispos ; 35(4): 237-52, 2014 May.
Article in English | MEDLINE | ID: mdl-24415392

ABSTRACT

(2R,3R,4S,5R)-2-(6-Amino-9H-purin-9-yl)-5-((((1r,3S)-3-(2-(5-(tert-butyl)-1H-benzo[d]imidazol-2-yl)ethyl)cyclobutyl)(isopropyl)amino)methyl)tetrahydrofuran-3,4-diol (EPZ-5676) is a novel DOT1L histone methyltransferase inhibitor currently in clinical development for the treatment of MLL-rearranged leukemias. This report describes the preclinical pharmacokinetics and metabolism of EPZ-5676, an aminonucleoside analog with exquisite target potency and selectivity that has shown robust and durable tumor growth inhibition in preclinical models. The in vivo pharmacokinetics in mouse, rat and dog were characterized following i.v. and p.o. administration; EPZ-5676 had moderate to high clearance, low oral bioavailability with a steady-state volume of distribution 2-3 fold higher than total body water. EPZ-5676 showed biexponential kinetics following i.v. administration, giving rise to a terminal elimination half-life (t1/2 ) of 1.1, 3.7 and 13.6 h in mouse, rat and dog, respectively. The corresponding in vitro ADME parameters were also studied and utilized for in vitro-in vivo extrapolation purposes. There was good agreement between the microsomal clearance and the in vivo clearance implicating hepatic oxidative metabolism as the predominant elimination route in preclinical species. Furthermore, low renal clearance was observed in mouse, approximating to fu -corrected glomerular filtration rate (GFR) and thus passive glomerular filtration. The metabolic pathways across species were studied in liver microsomes in which EPZ-5676 was metabolized to three monohydroxylated metabolites (M1, M3 and M5), one N-dealkylated product (M4) as well as an N-oxide (M6).


Subject(s)
Antineoplastic Agents/pharmacokinetics , Benzimidazoles/pharmacokinetics , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Methyltransferases/antagonists & inhibitors , Animals , Antineoplastic Agents/blood , Benzimidazoles/blood , Blood Proteins/metabolism , Dogs , Hepatocytes/metabolism , Madin Darby Canine Kidney Cells , Male , Mice , Microsomes, Liver/metabolism , Permeability , Rats, Sprague-Dawley
7.
Blood ; 122(6): 1017-25, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23801631

ABSTRACT

Rearrangements of the MLL gene define a genetically distinct subset of acute leukemias with poor prognosis. Current treatment options are of limited effectiveness; thus, there is a pressing need for new therapies for this disease. Genetic and small molecule inhibitor studies have demonstrated that the histone methyltransferase DOT1L is required for the development and maintenance of MLL-rearranged leukemia in model systems. Here we describe the characterization of EPZ-5676, a potent and selective aminonucleoside inhibitor of DOT1L histone methyltransferase activity. The compound has an inhibition constant value of 80 pM, and demonstrates 37 000-fold selectivity over all other methyltransferases tested. In cellular studies, EPZ-5676 inhibited H3K79 methylation and MLL-fusion target gene expression and demonstrated potent cell killing that was selective for acute leukemia lines bearing MLL translocations. Continuous IV infusion of EPZ-5676 in a rat xenograft model of MLL-rearranged leukemia caused complete tumor regressions that were sustained well beyond the compound infusion period with no significant weight loss or signs of toxicity. EPZ-5676 is therefore a potential treatment of MLL-rearranged leukemia and is under clinical investigation.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Leukemia/genetics , Leukemia/therapy , Methyltransferases/antagonists & inhibitors , Myeloid-Lymphoid Leukemia Protein/genetics , Animals , Cell Line, Tumor , Cell Proliferation , DNA Methylation , Dose-Response Relationship, Drug , Female , Histone Methyltransferases , Histones/metabolism , Humans , Neoplasm Transplantation , Protein Conformation , Rats , Rats, Nude
8.
Proc Natl Acad Sci U S A ; 110(19): 7922-7, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23620515

ABSTRACT

Inactivation of the switch/sucrose nonfermentable complex component SMARCB1 is extremely prevalent in pediatric malignant rhabdoid tumors (MRTs) or atypical teratoid rhabdoid tumors. This alteration is hypothesized to confer oncogenic dependency on EZH2 in these cancers. We report the discovery of a potent, selective, and orally bioavailable small-molecule inhibitor of EZH2 enzymatic activity, (N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4'-(morpholinomethyl)-[1,1'-biphenyl]-3-carboxamide). The compound induces apoptosis and differentiation specifically in SMARCB1-deleted MRT cells. Treatment of xenograft-bearing mice with (N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4'-(morpholinomethyl)-[1,1'-biphenyl]-3-carboxamide) leads to dose-dependent regression of MRTs with correlative diminution of intratumoral trimethylation levels of lysine 27 on histone H3, and prevention of tumor regrowth after dosing cessation. These data demonstrate the dependency of SMARCB1 mutant MRTs on EZH2 enzymatic activity and portend the utility of EZH2-targeted drugs for the treatment of these genetically defined cancers.


Subject(s)
Apoptosis , Neoplasms/therapy , Polycomb Repressive Complex 2/antagonists & inhibitors , Rhabdoid Tumor/enzymology , Rhabdoid Tumor/genetics , Animals , Antineoplastic Agents/pharmacology , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Cell Proliferation , Drug Design , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Gene Expression Profiling , HEK293 Cells , Histones/metabolism , Humans , Mice , Neoplasm Transplantation , Neoplasms/drug therapy , Neoplasms/pathology , Pyridines/pharmacology
9.
Nat Chem Biol ; 8(11): 890-6, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23023262

ABSTRACT

EZH2 catalyzes trimethylation of histone H3 lysine 27 (H3K27). Point mutations of EZH2 at Tyr641 and Ala677 occur in subpopulations of non-Hodgkin's lymphoma, where they drive H3K27 hypertrimethylation. Here we report the discovery of EPZ005687, a potent inhibitor of EZH2 (K(i) of 24 nM). EPZ005687 has greater than 500-fold selectivity against 15 other protein methyltransferases and has 50-fold selectivity against the closely related enzyme EZH1. The compound reduces H3K27 methylation in various lymphoma cells; this translates into apoptotic cell killing in heterozygous Tyr641 or Ala677 mutant cells, with minimal effects on the proliferation of wild-type cells. These data suggest that genetic alteration of EZH2 (for example, mutations at Tyr641 or Ala677) results in a critical dependency on enzymatic activity for proliferation (that is, the equivalent of oncogene addiction), thus portending the clinical use of EZH2 inhibitors for cancers in which EZH2 is genetically altered.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Histones/metabolism , Indazoles/pharmacology , Lymphoma/drug therapy , Lymphoma/pathology , Polycomb Repressive Complex 2/antagonists & inhibitors , Pyridones/pharmacology , Antineoplastic Agents/chemistry , Cell Cycle/drug effects , Cell Death/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Enhancer of Zeste Homolog 2 Protein , Enzyme Inhibitors/chemistry , Histones/chemistry , Humans , Indazoles/chemistry , Lymphoma/enzymology , Lymphoma/genetics , Lysine/metabolism , Methylation/drug effects , Molecular Structure , Point Mutation , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Pyridones/chemistry , Structure-Activity Relationship
10.
Cell Signal ; 23(3): 594-601, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21118717

ABSTRACT

The Schizosaccharomyces pombe fbp1 gene is transcriptionally repressed by protein kinase A (PKA) that is activated by extracellular glucose via a cAMP-signaling pathway. We previously used an fbp1-ura4 reporter that places uracil biosynthesis under the control of the glucose-sensing pathway to identify mutations in genes of the cAMP pathway. More recently, this reporter has been used in high throughput screens for small molecule inhibitors of heterologously-expressed cyclic nucleotide phosphodiesterases (PDEs) that hydrolyse cAMP to 5' AMP. Here we show that strains lacking the adenylyl cyclase gene respond to either exogenous cAMP or cGMP to activate PKA, thus regulating fbp1-ura4 expression and other PKA-regulated processes such as conjugation and the nuclear export of an Rst2-GFP fusion protein. Expression of cGMP-specific PDEs or ones that hydrolyse both cAMP and cGMP increases the amount of exogenous cGMP required to activate PKA in order to repress fbp1-ura4 expression, creating conditions that allow detection of inhibitors of these PDEs. As proof of this concept, we screened a collection of compounds previously identified as inhibitors of cAMP-specific PDE4 or PDE7 enzymes for their ability to inhibit the mammalian cGMP-specific PDE5A enzyme. We identified compound BC76, which inhibits PDE5A in an in vitro enzyme assay with an IC(50) of 232nM. Further yeast-based assays show that BC76 inhibits PDE1, PDE4, PDE5, PDE8, PDE10 and PDE11, thus demonstrating the utility of this system for detecting and characterising inhibitors of either cAMP- or cGMP-metabolising PDEs.


Subject(s)
Benzimidazoles/pharmacology , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Drug Evaluation, Preclinical/methods , Genes, Reporter , Phosphodiesterase 5 Inhibitors/pharmacology , Pyrroles/pharmacology , Recombinant Proteins/biosynthesis , Schizosaccharomyces/genetics , Animals , Cattle , Humans , Mice
11.
J Biomol Screen ; 15(4): 359-67, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20228279

ABSTRACT

Studies of the phosphodiesterase PDE7 family are impeded by there being only one commercially available PDE7 inhibitor, BRL50481. The authors have employed a high-throughput screen of commercial chemical libraries, using a fission yeast-based assay, to identify PDE7 inhibitors that include steroids, podocarpanes, and an unusual heterocyclic compound, BC30. In vitro enzyme assays measuring the potency of BC30 and 2 podocarpanes, in comparison with BRL50481, produce data consistent with those from yeast-based assays. In other enzyme assays, BC30 stimulates the PDE4D catalytic domain but not full-length PDE4D2, suggesting an allosteric site of action. BC30 significantly enhances the anti-inflammatory effect of the PDE4 inhibitor rolipram as measured by release of tumor necrosis factor alpha from activated monocytes. These studies introduce several new PDE7 inhibitors that may be excellent candidates for medicinal chemistry because of the requirements for drug-like characteristics placed on them by the nature of the yeast-based screen.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 7/antagonists & inhibitors , High-Throughput Screening Assays/methods , Phosphodiesterase Inhibitors/analysis , Phosphodiesterase Inhibitors/pharmacology , Schizosaccharomyces/metabolism , Anti-Inflammatory Agents/pharmacology , Catalytic Domain , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 7/chemistry , Enzyme Assays , Humans , Hydrolysis/drug effects , Inhibitory Concentration 50 , Lipopolysaccharides/pharmacology , Orotic Acid/analogs & derivatives , Orotic Acid/pharmacology , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/classification , Schizosaccharomyces/drug effects , Schizosaccharomyces/growth & development , Tumor Necrosis Factor-alpha/metabolism
12.
J Biomol Screen ; 13(1): 62-71, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18227226

ABSTRACT

Cyclic nucleotide phosphodiesterases (PDEs) comprise a superfamily of enzymes that serve as drug targets in many human diseases. There is a continuing need to identify high-specificity inhibitors that affect individual PDE families or even subtypes within a single family. The authors describe a fission yeast-based high-throughput screen to detect inhibitors of heterologously expressed adenosine 3',5'-cyclic monophosphate (cAMP) PDEs. The utility of this system is demonstrated by the construction and characterization of strains that express mammalian PDE2A, PDE4A, PDE4B, and PDE8A and respond appropriately to known PDE2A and PDE4 inhibitors. High-throughput screens of 2 bioactive compound libraries for PDE inhibitors using strains expressing PDE2A, PDE4A, PDE4B, and the yeast PDE Cgs2 identified known PDE inhibitors and members of compound classes associated with PDE inhibition. The authors verified that the furanocoumarin imperatorin is a PDE4 inhibitor based on its ability to produce a PDE4-specific elevation of cAMP levels. This platform can be used to identify PDE activators, as well as genes encoding PDE regulators, which could serve as targets for future drug screens.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Phosphodiesterase Inhibitors/pharmacology , Schizosaccharomyces/drug effects , Schizosaccharomyces/enzymology , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Animals , Cyclic AMP/metabolism , Enzyme Activation/drug effects , Genes, Fungal , Genes, Reporter , Mice , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Schizosaccharomyces/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...