Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Cancer Cell ; 39(9): 1214-1226.e10, 2021 09 13.
Article in English | MEDLINE | ID: mdl-34375612

ABSTRACT

PARP7 is a monoPARP that catalyzes the transfer of single units of ADP-ribose onto substrates to change their function. Here, we identify PARP7 as a negative regulator of nucleic acid sensing in tumor cells. Inhibition of PARP7 restores type I interferon (IFN) signaling responses to nucleic acids in tumor models. Restored signaling can directly inhibit cell proliferation and activate the immune system, both of which contribute to tumor regression. Oral dosing of the PARP7 small-molecule inhibitor, RBN-2397, results in complete tumor regression in a lung cancer xenograft and induces tumor-specific adaptive immune memory in an immunocompetent mouse cancer model, dependent on inducing type I IFN signaling in tumor cells. PARP7 is a therapeutic target whose inhibition induces both cancer cell-autonomous and immune stimulatory effects via enhanced IFN signaling. These data support the targeting of a monoPARP in cancer and introduce a potent and selective PARP7 inhibitor to enter clinical development.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Interferon Type I/metabolism , Neoplasms/drug therapy , Nucleoside Transport Proteins/genetics , Nucleoside Transport Proteins/metabolism , Small Molecule Libraries/administration & dosage , Adaptive Immunity/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , HEK293 Cells , HeLa Cells , Humans , Mice , Neoplasms/genetics , Neoplasms/metabolism , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Tumor Escape/drug effects , Xenograft Model Antitumor Assays
2.
Chembiochem ; 22(12): 2107-2110, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33838082

ABSTRACT

PARP14 is an interferon-stimulated gene that is overexpressed in multiple tumor types, influencing pro-tumor macrophage polarization as well as suppressing the antitumor inflammation response by modulating IFN-γ and IL-4 signaling. PARP14 is a 203 kDa protein that possesses a catalytic domain responsible for the transfer of mono-ADP-ribose to its substrates. PARP14 also contains three macrodomains and a WWE domain which are binding modules for mono-ADP-ribose and poly-ADP-ribose, respectively, in addition to two RNA recognition motifs. Catalytic inhibitors of PARP14 have been shown to reverse IL-4 driven pro-tumor gene expression in macrophages, however it is not clear what roles the non-enzymatic biomolecular recognition motifs play in PARP14-driven immunology and inflammation. To further understand this, we have discovered a heterobifunctional small molecule designed based on a catalytic inhibitor of PARP14 that binds in the enzyme's NAD+ -binding site and recruits cereblon to ubiquitinate it and selectively target it for degradation.


Subject(s)
Poly(ADP-ribose) Polymerases/metabolism , Small Molecule Libraries/pharmacology , Humans , Macrophages/drug effects , Macrophages/metabolism , Molecular Structure , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry
3.
Cell Chem Biol ; 28(8): 1158-1168.e13, 2021 08 19.
Article in English | MEDLINE | ID: mdl-33705687

ABSTRACT

PARP14 has been implicated by genetic knockout studies to promote protumor macrophage polarization and suppress the antitumor inflammatory response due to its role in modulating interleukin-4 (IL-4) and interferon-γ signaling pathways. Here, we describe structure-based design efforts leading to the discovery of a potent and highly selective PARP14 chemical probe. RBN012759 inhibits PARP14 with a biochemical half-maximal inhibitory concentration of 0.003 µM, exhibits >300-fold selectivity over all PARP family members, and its profile enables further study of PARP14 biology and disease association both in vitro and in vivo. Inhibition of PARP14 with RBN012759 reverses IL-4-driven protumor gene expression in macrophages and induces an inflammatory mRNA signature similar to that induced by immune checkpoint inhibitor therapy in primary human tumor explants. These data support an immune suppressive role of PARP14 in tumors and suggest potential utility of PARP14 inhibitors in the treatment of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Inflammation/drug therapy , Interleukin-4/antagonists & inhibitors , Kidney Neoplasms/drug therapy , Macrophages/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Dose-Response Relationship, Drug , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Inflammation/genetics , Inflammation/metabolism , Interleukin-4/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Poly(ADP-ribose) Polymerases/genetics , RAW 264.7 Cells , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
Cell Chem Biol ; 27(7): 877-887.e14, 2020 07 16.
Article in English | MEDLINE | ID: mdl-32679093

ABSTRACT

Poly(ADP-ribose) polymerase (PARP) enzymes use nicotinamide adenine dinucleotide (NAD+) to modify up to seven different amino acids with a single mono(ADP-ribose) unit (MARylation deposited by PARP monoenzymes) or branched poly(ADP-ribose) polymers (PARylation deposited by PARP polyenzymes). To enable the development of tool compounds for PARP monoenzymes and polyenzymes, we have developed active site probes for use in in vitro and cellular biophysical assays to characterize active site-directed inhibitors that compete for NAD+ binding. These assays are agnostic of the protein substrate for each PARP, overcoming a general lack of knowledge around the substrates for these enzymes. The in vitro assays use less enzyme than previously described activity assays, enabling discrimination of inhibitor potencies in the single-digit nanomolar range, and the cell-based assays can differentiate compounds with sub-nanomolar potencies and measure inhibitor residence time in live cells.


Subject(s)
Fluorescent Dyes/chemistry , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Poly(ADP-ribose) Polymerases/metabolism , Binding, Competitive , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/metabolism , HEK293 Cells , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , NAD/chemistry , NAD/metabolism , Nanoparticles/chemistry , Poly(ADP-ribose) Polymerase Inhibitors/chemical synthesis , Poly(ADP-ribose) Polymerase Inhibitors/metabolism , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/genetics , Protein Binding , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Surface Plasmon Resonance
5.
SLAS Discov ; 25(3): 241-252, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31855104

ABSTRACT

Mono(ADP-ribosylation) (MARylation) and poly(ADP-ribosylation) (PARylation) are posttranslational modifications found on multiple amino acids. There are 12 enzymatically active mono(ADP-ribose) polymerase (monoPARP) enzymes and 4 enzymatically active poly(ADP-ribose) polymerase (polyPARP) enzymes that use nicotinamide adenine dinucleotide (NAD+) as the ADP-ribose donating substrate to generate these modifications. While there are approved drugs and clinical trials ongoing for the enzymes that perform PARylation, MARylation is gaining recognition for its role in immune function, inflammation, and cancer. However, there is a lack of chemical probes to study the function of monoPARPs in cells and in vivo. An important first step to generating chemical probes for monoPARPs is to develop biochemical assays to enable hit finding, and determination of the potency and selectivity of inhibitors. Complicating the development of enzymatic assays is that it is poorly understood how monoPARPs engage their substrates. To overcome this, we have developed a family-wide approach to developing robust high-throughput monoPARP assays where the enzymes are immobilized and forced to self-modify using biotinylated-NAD+, which is detected using a dissociation-enhanced lanthanide fluorescence immunoassay (DELFIA) readout. Herein we describe the development of assays for 12 monoPARPs and 3 polyPARPs and apply them to understand the potency and selectivity of a focused library of inhibitors across this family.


Subject(s)
ADP Ribose Transferases/antagonists & inhibitors , Enzyme Inhibitors/isolation & purification , High-Throughput Screening Assays , Poly(ADP-ribose) Polymerase Inhibitors/isolation & purification , Protein Processing, Post-Translational/genetics , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/genetics , ADP-Ribosylation/genetics , Adenosine Diphosphate Ribose/genetics , Enzyme Inhibitors/pharmacology , Humans , NAD/chemistry , Poly ADP Ribosylation/genetics , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Poly(ADP-ribose) Polymerases/drug effects , Poly(ADP-ribose) Polymerases/genetics , Substrate Specificity
6.
PLoS One ; 13(6): e0197372, 2018.
Article in English | MEDLINE | ID: mdl-29856759

ABSTRACT

A key challenge in the development of precision medicine is defining the phenotypic consequences of pharmacological modulation of specific target macromolecules. To address this issue, a variety of genetic, molecular and chemical tools can be used. All of these approaches can produce misleading results if the specificity of the tools is not well understood and the proper controls are not performed. In this paper we illustrate these general themes by providing detailed studies of small molecule inhibitors of the enzymatic activity of two members of the SMYD branch of the protein lysine methyltransferases, SMYD2 and SMYD3. We show that tool compounds as well as CRISPR/Cas9 fail to reproduce many of the cell proliferation findings associated with SMYD2 and SMYD3 inhibition previously obtained with RNAi based approaches and with early stage chemical probes.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Carcinogenesis/genetics , Histone-Lysine N-Methyltransferase/genetics , A549 Cells , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , CRISPR-Cas Systems , Carcinogenesis/drug effects , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/chemistry , Humans , Methylation/drug effects , Methyltransferases/antagonists & inhibitors , RNA Interference , Small Molecule Libraries/pharmacology
7.
PLoS One ; 13(5): e0197082, 2018.
Article in English | MEDLINE | ID: mdl-29742153

ABSTRACT

WHSC1 is a histone methyltransferase that is responsible for mono- and dimethylation of lysine 36 on histone H3 and has been implicated as a driver in a variety of hematological and solid tumors. Currently, there is a complete lack of validated chemical matter for this important drug discovery target. Herein we report on the first fully validated WHSC1 inhibitor, PTD2, a norleucine-containing peptide derived from the histone H4 sequence. This peptide exhibits micromolar affinity towards WHSC1 in biochemical and biophysical assays. Furthermore, a crystal structure was solved with the peptide in complex with SAM and the SET domain of WHSC1L1. This inhibitor is an important first step in creating potent, selective WHSC1 tool compounds for the purposes of understanding the complex biology in relation to human disease.


Subject(s)
Enzyme Inhibitors/chemistry , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Neoplasms/drug therapy , Peptides/chemistry , Repressor Proteins/antagonists & inhibitors , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Histones/chemistry , Histones/genetics , Humans , Lysine/chemistry , Neoplasms/enzymology , Norleucine/analogs & derivatives , Norleucine/chemistry , Norleucine/pharmacology , PR-SET Domains/genetics , Peptides/genetics , Protein Conformation/drug effects , Repressor Proteins/chemistry , Repressor Proteins/genetics
9.
ACS Med Chem Lett ; 7(2): 134-8, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26985287

ABSTRACT

SMYD3 has been implicated in a range of cancers; however, until now no potent selective small molecule inhibitors have been available for target validation studies. A novel oxindole series of SMYD3 inhibitors was identified through screening of the Epizyme proprietary histone methyltransferase-biased library. Potency optimization afforded two tool compounds, sulfonamide EPZ031686 and sulfamide EPZ030456, with cellular potency at a level sufficient to probe the in vitro biology of SMYD3 inhibition. EPZ031686 shows good bioavailability following oral dosing in mice making it a suitable tool for potential in vivo target validation studies.

10.
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
11.
ACS Med Chem Lett ; 6(5): 491-5, 2015 May 14.
Article in English | MEDLINE | ID: mdl-26005520

ABSTRACT

Inhibitors of the protein methyltransferase Enhancer of Zeste Homolog 2 (EZH2) may have significant therapeutic potential for the treatment of B cell lymphomas and other cancer indications. The ability of the scientific community to explore fully the spectrum of EZH2-associated pathobiology has been hampered by the lack of in vivo-active tool compounds for this enzyme. Here we report the discovery and characterization of EPZ011989, a potent, selective, orally bioavailable inhibitor of EZH2 with useful pharmacokinetic properties. EPZ011989 demonstrates significant tumor growth inhibition in a mouse xenograft model of human B cell lymphoma. Hence, this compound represents a powerful tool for the expanded exploration of EZH2 activity in biology.

12.
J Biomol Screen ; 20(6): 810-20, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25755264

ABSTRACT

Demethylation of histones by lysine demethylases (KDMs) plays a critical role in controlling gene transcription. Aberrant demethylation may play a causal role in diseases such as cancer. Despite the biological significance of these enzymes, there are limited assay technologies for study of KDMs and few quality chemical probes available to interrogate their biology. In this report, we demonstrate the utility of self-assembled monolayer desorption/ionization (SAMDI) mass spectrometry for the investigation of quantitative KDM enzyme kinetics and for high-throughput screening for KDM inhibitors. SAMDI can be performed in 384-well format and rapidly allows reaction components to be purified prior to injection into a mass spectrometer, without a throughput-limiting liquid chromatography step. We developed sensitive and robust assays for KDM1A (LSD1, AOF2) and KDM4C (JMJD2C, GASC1) and screened 13,824 compounds against each enzyme. Hits were rapidly triaged using a redox assay to identify compounds that interfered with the catalytic oxidation chemistry used by the KDMs for the demethylation reaction. We find that overall this high-throughput mass spectrometry platform coupled with the elimination of redox active compounds leads to a hit rate that is manageable for follow-up work.


Subject(s)
Artifacts , High-Throughput Screening Assays/methods , Histone Demethylases/metabolism , Mass Spectrometry/methods , Oxidation-Reduction , Drug Discovery/methods , Enzyme Assays , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Methylation , Small Molecule Libraries
13.
PLoS One ; 9(12): e111840, 2014.
Article in English | MEDLINE | ID: mdl-25493630

ABSTRACT

Patients with non-Hodgkin lymphoma (NHL) are treated today with a cocktail of drugs referred to as CHOP (Cyclophosphamide, Hydroxyldaunorubicin, Oncovin, and Prednisone). Subsets of patients with NHL of germinal center origin bear oncogenic mutations in the EZH2 histone methyltransferase. Clinical testing of the EZH2 inhibitor EPZ-6438 has recently begun in patients. We report here that combining EPZ-6438 with CHOP in preclinical cell culture and mouse models results in dramatic synergy for cell killing in EZH2 mutant germinal center NHL cells. Surprisingly, we observe that much of this synergy is due to Prednisolone - a glucocorticoid receptor agonist (GRag) component of CHOP. Dramatic synergy was observed when EPZ-6438 is combined with Prednisolone alone, and a similar effect was observed with Dexamethasone, another GRag. Remarkably, the anti-proliferative effect of the EPZ-6438+GRag combination extends beyond EZH2 mutant-bearing cells to more generally impact germinal center NHL. These preclinical data reveal an unanticipated biological intersection between GR-mediated gene regulation and EZH2-mediated chromatin remodeling. The data also suggest the possibility of a significant and practical benefit of combining EZH2 inhibitors and GRag that warrants further investigation in a clinical setting.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzamides/pharmacology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Lymphoma, Non-Hodgkin/drug therapy , Pyridones/pharmacology , Animals , Biphenyl Compounds , Cell Line, Tumor , Cyclophosphamide/pharmacology , Dexamethasone/pharmacology , Doxorubicin/pharmacology , Drug Evaluation, Preclinical , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Glucocorticoids/pharmacology , Humans , Lymphoma, Non-Hodgkin/metabolism , Mice, SCID , Morpholines , Neoplasm Transplantation , Prednisolone/pharmacology , Prednisone/pharmacology , Random Allocation , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/metabolism , Vincristine/pharmacology
14.
ACS Chem Biol ; 9(11): 2459-64, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25154026

ABSTRACT

EZH2 and EZH1 are protein methyltransferases (PMTs) responsible for histone H3, lysine 27 (H3K27) methylation. Trimethylation of H3K27 (H3K27me3) is a hallmark of many cancers, including non-Hodgkin lymphoma (NHL). Heterozygous EZH2 point mutations at Tyr641, Ala677, and Ala687 have been observed in NHL. The Tyr641 mutations enhance activity on H3K27me2 but have weak or no activity on unmethylated H3K27, whereas the Ala677 and Ala687 mutations use substrates of all methylation states effectively. It has been proposed that enzymatic coupling of the wild-type and mutant enzymes leads to the oncogenic H3K27me3 mark in mutant-bearing NHL. We show that coupling with the wild-type enzyme is needed to achieve H3K27me3 for several mutants, but that others are capable of achieving H3K27me3 on their own. All forms of PRC2 (wild-type and mutants) display kinetic signatures that are consistent with a distributive mechanism of catalysis.


Subject(s)
Mutation , Neoplasms/genetics , Polycomb Repressive Complex 2/chemistry , Enhancer of Zeste Homolog 2 Protein , Humans , Polycomb Repressive Complex 2/genetics
15.
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
16.
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
17.
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
18.
Chem Biol Drug Des ; 80(6): 971-80, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22978415

ABSTRACT

DOT1L is the human protein methyltransferase responsible for catalyzing the methylation of histone H3 on lysine 79 (H3K79). The ectopic activity of DOT1L, associated with the chromosomal translocation that is a universal hallmark of MLL-rearranged leukemia, is a required driver of leukemogenesis in this malignancy. Here, we present studies on the structure-activity relationship of aminonucleoside-based DOT1L inhibitors. Within this series, we find that improvements in target enzyme affinity and selectivity are driven entirely by diminution of the dissociation rate constant for the enzyme-inhibitor complex, leading to long residence times for the binary complex. The biochemical K(i) and residence times measured for these inhibitors correlate well with their effects on intracellular H3K79 methylation and MLL-rearranged leukemic cell killing. Crystallographic studies reveal a conformational adaptation mechanism associated with high-affinity inhibitor binding and prolonged residence time; these studies also suggest that conformational adaptation likewise plays a critical role in natural ligand interactions with the enzyme, hence, facilitating enzyme turnover. These results provide critical insights into the role of conformational adaptation in the enzymatic mechanism of catalysis and in pharmacologic intervention for DOT1L and other members of this enzyme class.


Subject(s)
Enzyme Inhibitors/chemistry , Methyltransferases/antagonists & inhibitors , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/metabolism , Histone-Lysine N-Methyltransferase , Histones/metabolism , Humans , Kinetics , Methylation , Methyltransferases/metabolism , Molecular Docking Simulation , Nucleosides/chemistry , Nucleosides/metabolism , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship
19.
FEBS Lett ; 586(19): 3448-51, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22850114

ABSTRACT

Heterozygous point mutations at Y641 and A677 in the EZH2 SET domain are prevalent in about 10-24% of Non-Hodgkin lymphomas (NHL). Previous studies indicate that these are gain-of-function mutations leading to the hypertrimethylation of H3K27. These EZH2 mutations may drive the proliferation of lymphoma and make EZH2 a molecular target for patients harboring these mutations. Here, another EZH2 SET domain point mutation, A687V, occurring in about 1-2% of lymphoma patients, is also shown to be a gain-of-function mutation that greatly enhances its ability to perform dimethylation relative to wild-type EZH2 and is equally proficient at catalyzing trimethylation. We propose that A687V EZH2 also leads to hypertrimethylation of H3K27 and may thus be a driver mutation in NHL.


Subject(s)
Lymphoma, Non-Hodgkin/enzymology , Lymphoma, Non-Hodgkin/genetics , Mutant Proteins/genetics , Neoplasm Proteins/genetics , Point Mutation , Polycomb Repressive Complex 2/genetics , Enhancer of Zeste Homolog 2 Protein , Heterozygote , Histones/chemistry , Histones/metabolism , Humans , Kinetics , Methylation , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
20.
FEBS Lett ; 585(19): 3011-4, 2011 Oct 03.
Article in English | MEDLINE | ID: mdl-21856302

ABSTRACT

Mutations at tyrosine 641 (Y641F, Y641N, Y641S and Y641H) in the SET domain of EZH2 have been identified in patients with certain subtypes of non-Hodgkin lymphoma (NHL). These mutations were shown to change the substrate specificity of EZH2 for various methylation states of lysine 27 on histone H3 (H3K27). An additional mutation at EZH2 Y641 to cysteine (Y641C) was also found in one patient with NHL and in SKM-1 cells derived from a patient with myelodisplastic syndrome (MDS). The Y641C mutation has been reported to dramatically reduce enzymatic activity. Here, we demonstrate that while the Y641C mutation ablates enzymatic activity against unmethylated and monomethylated H3K27, it is superior to wild-type in catalyzing the formation of trimethylated H3K27 from the dimethylated precursor.


Subject(s)
DNA-Binding Proteins/metabolism , Histones/metabolism , Lysine/metabolism , Mutation , Substrate Specificity/genetics , Transcription Factors/metabolism , Animals , Base Sequence , Cell Line , Cysteine/chemistry , Cysteine/metabolism , DNA-Binding Proteins/genetics , Enhancer of Zeste Homolog 2 Protein , Histones/genetics , Humans , Lysine/chemistry , Methylation , Molecular Sequence Data , Polycomb Repressive Complex 2 , Sequence Analysis, DNA , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL