Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Biochemistry ; 62(14): 2147-2160, 2023 07 18.
Article in English | MEDLINE | ID: mdl-37403936

ABSTRACT

Werner syndrome protein (WRN) is a multifunctional enzyme with helicase, ATPase, and exonuclease activities that are necessary for numerous DNA-related transactions in the human cell. Recent studies identified WRN as a synthetic lethal target in cancers characterized by genomic microsatellite instability resulting from defects in DNA mismatch repair pathways. WRN's helicase activity is essential for the viability of these high microsatellite instability (MSI-H) cancers and thus presents a therapeutic opportunity. To this end, we developed a multiplexed high-throughput screening assay that monitors exonuclease, ATPase, and helicase activities of full-length WRN. This screening campaign led to the discovery of 2-sulfonyl/sulfonamide pyrimidine derivatives as novel covalent inhibitors of WRN helicase activity. The compounds are specific for WRN versus other human RecQ family members and show competitive behavior with ATP. Examination of these novel chemical probes established the sulfonamide NH group as a key driver of compound potency. One of the leading compounds, H3B-960, showed consistent activities in a range of assays (IC50 = 22 nM, KD = 40 nM, KI = 32 nM), and the most potent compound identified, H3B-968, has inhibitory activity IC50 ∼ 10 nM. These kinetic properties trend toward other known covalent druglike molecules. Our work provides a new avenue for screening WRN for inhibitors that may be adaptable to different therapeutic modalities such as targeted protein degradation, as well as a proof of concept for the inhibition of WRN helicase activity by covalent molecules.


Subject(s)
Neoplasms , Werner Syndrome , Humans , Exodeoxyribonucleases/genetics , RecQ Helicases/genetics , RecQ Helicases/metabolism , High-Throughput Screening Assays , Microsatellite Instability , Werner Syndrome Helicase/metabolism
2.
J Biol Chem ; 298(11): 102539, 2022 11.
Article in English | MEDLINE | ID: mdl-36179791

ABSTRACT

Recent studies have reported that the peroxisome proliferator-activated receptor gamma (PPARγ) pathway is activated in approximately 40% of patients with muscle-invasive bladder cancer. This led us to investigate pharmacological repression of PPARγ as a possible intervention strategy. Here, we characterize PPARγ antagonists and inverse agonists and find that the former behave as silent ligands, whereas inverse agonists (T0070907 and SR10221) repress downstream PPARγ target genes leading to growth inhibition in bladder cancer cell lines. To understand the mechanism, we determined the ternary crystal structure of PPARγ bound to T0070907 and the corepressor (co-R) peptide NCOR1. The structure shows that the AF-2 helix 12 (H12) rearranges to bind inside the ligand-binding domain, where it forms stabilizing interactions with the compound. This dramatic movement in H12 unveils a large interface for co-R binding. In contrast, the crystal structure of PPARγ bound to a SR10221 analog shows more subtle structural differences, where the compound binds and pushes H12 away from the ligand-binding domain to allow co-R binding. Interestingly, we found that both classes of compound promote recruitment of co-R proteins in biochemical assays but with distinct conformational changes in H12. We validate our structural models using both site-directed mutagenesis and chemical probes. Our findings offer new mechanistic insights into pharmacological modulation of PPARγ signaling.


Subject(s)
PPAR gamma , Urinary Bladder Neoplasms , Humans , PPAR gamma/metabolism , Ligands , Benzamides/pharmacology
3.
Nat Commun ; 12(1): 4491, 2021 07 23.
Article in English | MEDLINE | ID: mdl-34301950

ABSTRACT

Intron selection during the formation of prespliceosomes is a critical event in pre-mRNA splicing. Chemical modulation of intron selection has emerged as a route for cancer therapy. Splicing modulators alter the splicing patterns in cells by binding to the U2 snRNP (small nuclear ribonucleoprotein)-a complex chaperoning the selection of branch and 3' splice sites. Here we report crystal structures of the SF3B module of the U2 snRNP in complex with spliceostatin and sudemycin FR901464 analogs, and the cryo-electron microscopy structure of a cross-exon prespliceosome-like complex arrested with spliceostatin A. The structures reveal how modulators inactivate the branch site in a sequence-dependent manner and stall an E-to-A prespliceosome intermediate by covalent coupling to a nucleophilic zinc finger belonging to the SF3B subunit PHF5A. These findings support a mechanism of intron recognition by the U2 snRNP as a toehold-mediated strand invasion and advance an unanticipated drug targeting concept.


Subject(s)
DNA/genetics , Introns/genetics , Pyrans/metabolism , Ribonucleoprotein, U2 Small Nuclear/metabolism , Spiro Compounds/metabolism , Spliceosomes/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , DNA/chemistry , DNA/metabolism , Humans , Lactones/chemistry , Lactones/metabolism , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Protein Conformation , Pyrans/chemistry , Pyrones/chemistry , Pyrones/metabolism , Ribonucleoprotein, U2 Small Nuclear/chemistry , Spiro Compounds/chemistry , Spliceosomes/ultrastructure
4.
ACS Med Chem Lett ; 12(1): 93-98, 2021 Jan 14.
Article in English | MEDLINE | ID: mdl-33488969

ABSTRACT

Fibroblast growth factor receptors (FGFR) 2 and 3 have been established as drivers of numerous types of cancer with multiple drugs approved or entering late stage clinical trials. A limitation of current inhibitors is vulnerability to gatekeeper resistance mutations. Using a combination of targeted high-throughput screening and structure-based drug design, we have developed a series of aminopyrazole based FGFR inhibitors that covalently target a cysteine residue on the P-loop of the kinase. The inhibitors show excellent activity against the wild-type and gatekeeper mutant versions of the enzymes. Further optimization using SAR analysis and structure-based drug design led to analogues with improved potency and drug metabolism and pharmacokinetics properties.

5.
Subcell Biochem ; 96: 409-432, 2021.
Article in English | MEDLINE | ID: mdl-33252738

ABSTRACT

In this chapter, the essential role of the SF3b multi-protein complex will be discussed in the context of the overall spliceosome. SF3b is critical during spliceosome assembly for recognition of the branch point (BP) adenosine and, by de facto, selection of the 3' splice site. This complex is highly dynamic, undergoing significant conformational changes upon loading of the branch duplex RNA and in its relative positioning during spliceosomal remodeling from the A, pre-B, B, Bact and B* complexes. Ultimately, during the spliceosome activation phase, SF3b must be displaced to unmask the branch point adenosine for the first splicing reaction to occur. In certain cancers, such as the hematological malignancies CML, CLL and MDS, the SF3b subunit SF3B1 is frequently mutated. Recent studies suggest these mutations lead to inappropriate branch point selection and mis-splicing events that appear to be drivers of disease. Finally, the SF3b complex is the target for at least three different classes of natural product-based inhibitors. These inhibitors bind in the BP adenosine-binding pocket and demonstrate a pre-mRNA competitive mechanism of action resulting in either intron retention or exon skipping. These compounds are extremely useful as chemical probes to isolate and characterize early stages of spliceosome assembly. They are also being explored preclinically and clinically as possible agents for hematological cancers.


Subject(s)
Biological Products/pharmacology , Multiprotein Complexes/antagonists & inhibitors , Spliceosomes/chemistry , Spliceosomes/drug effects , Hematologic Neoplasms/drug therapy , Humans , Multiprotein Complexes/genetics , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing , Spliceosomes/genetics , Spliceosomes/metabolism
6.
ACS Med Chem Lett ; 11(6): 1305-1309, 2020 Jun 11.
Article in English | MEDLINE | ID: mdl-32551016

ABSTRACT

Carbamoyl phosphate synthetase 1 (CPS1) is a potential synthetic lethal target in LKB1-deficient nonsmall cell lung cancer, where its overexpression supports the production of pyrimidine synthesis. In other cancer types, CPS1 overexpression and activity may prevent the accumulation of toxic levels of intratumoral ammonia to support tumor growth. Herein we report the discovery of a novel series of potent and selective small-molecule inhibitors of CPS1. Piperazine 2 was initially identified as a promising CPS1 inhibitor through a high-throughput screening effort. Subsequent structure-activity relationship optimization and structure-based drug design led to the discovery of piperazine H3B-616 (25), a potent allosteric inhibitor of CPS1 (IC50 = 66 nM).

7.
Cell Chem Biol ; 27(3): 259-268.e5, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32017919

ABSTRACT

Carbamoyl phosphate synthetase 1 (CPS1) catalyzes the first step in the ammonia-detoxifying urea cycle, converting ammonia to carbamoyl phosphate under physiologic conditions. In cancer, CPS1 overexpression supports pyrimidine synthesis to promote tumor growth in some cancer types, while in others CPS1 activity prevents the buildup of toxic levels of intratumoral ammonia to allow for sustained tumor growth. Targeted CPS1 inhibitors may, therefore, provide a therapeutic benefit for cancer patients with tumors overexpressing CPS1. Herein, we describe the discovery of small-molecule CPS1 inhibitors that bind to a previously unknown allosteric pocket to block ATP hydrolysis in the first step of carbamoyl phosphate synthesis. CPS1 inhibitors are active in cellular assays, blocking both urea synthesis and CPS1 support of the pyrimidine biosynthetic pathway, while having no activity against CPS2. These newly discovered CPS1 inhibitors are a first step toward providing researchers with valuable tools for probing CPS1 cancer biology.


Subject(s)
Carbamoyl-Phosphate Synthase (Ammonia)/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Piperidines/pharmacology , Small Molecule Libraries/pharmacology , Thiazoles/pharmacology , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Allosteric Regulation/drug effects , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Humans , Hydrolysis/drug effects , Models, Molecular , Molecular Structure , Piperidines/chemistry , Small Molecule Libraries/chemistry , Thiazoles/chemistry
8.
J Biol Chem ; 294(45): 16966-16977, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31582562

ABSTRACT

DNMT3A (DNA methyltransferase 3A) is a de novo DNA methyltransferase responsible for establishing CpG methylation patterns within the genome. DNMT3A activity is essential for normal development, and its dysfunction has been linked to developmental disorders and cancer. DNMT3A is frequently mutated in myeloid malignancies with the majority of mutations occurring at Arg-882, where R882H mutations are most frequent. The R882H mutation causes a reduction in DNA methyltransferase activity and hypomethylation at differentially-methylated regions within the genome, ultimately preventing hematopoietic stem cell differentiation and leading to leukemogenesis. Although the means by which the R882H DNMT3A mutation reduces enzymatic activity has been the subject of several studies, the precise mechanism by which this occurs has been elusive. Herein, we demonstrate that in the context of the full-length DNMT3A protein, the R882H mutation stabilizes the formation of large oligomeric DNMT3A species to reduce the overall DNA methyltransferase activity of the mutant protein as well as the WT-R882H complex in a dominant-negative manner. This shift in the DNMT3A oligomeric equilibrium and the resulting reduced enzymatic activity can be partially rescued in the presence of oligomer-disrupting DNMT3L, as well as DNMT3A point mutations along the oligomer-forming interface of the catalytic domain. In addition to modulating the oligomeric state of DNMT3A, the R882H mutation also leads to a DNA-binding defect, which may further reduce enzymatic activity. These findings provide a mechanistic explanation for the observed loss of DNMT3A activity associated with the R882H hot spot mutation in cancer.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA (Cytosine-5-)-Methyltransferases/metabolism , Mutation , Protein Multimerization , DNA/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Humans , Models, Molecular , Protein Structure, Quaternary
9.
Genes Dev ; 32(3-4): 309-320, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29491137

ABSTRACT

Somatic mutations in spliceosome proteins lead to dysregulated RNA splicing and are observed in a variety of cancers. These genetic aberrations may offer a potential intervention point for targeted therapeutics. SF3B1, part of the U2 small nuclear RNP (snRNP), is targeted by splicing modulators, including E7107, the first to enter clinical trials, and, more recently, H3B-8800. Modulating splicing represents a first-in-class opportunity in drug discovery, and elucidating the structural basis for the mode of action opens up new possibilities for structure-based drug design. Here, we present the cryogenic electron microscopy (cryo-EM) structure of the SF3b subcomplex (SF3B1, SF3B3, PHF5A, and SF3B5) bound to E7107 at 3.95 Å. This structure shows that E7107 binds in the branch point adenosine-binding pocket, forming close contacts with key residues that confer resistance upon mutation: SF3B1R1074H and PHF5AY36C The structure suggests a model in which splicing modulators interfere with branch point adenosine recognition and supports a substrate competitive mechanism of action (MOA). Using several related chemical probes, we validate the pose of the compound and support their substrate competitive MOA by comparing their activity against both strong and weak pre-mRNA substrates. Finally, we present functional data and structure-activity relationship (SAR) on the PHF5AR38C mutation that sensitizes cells to some chemical probes but not others. Developing small molecule splicing modulators represents a promising therapeutic approach for a variety of diseases, and this work provides a significant step in enabling structure-based drug design for these elaborate natural products. Importantly, this work also demonstrates that the utilization of cryo-EM in drug discovery is coming of age.


Subject(s)
Epoxy Compounds/chemistry , Macrolides/chemistry , Phosphoproteins/chemistry , RNA Splicing Factors/chemistry , RNA Splicing/drug effects , Spliceosomes/drug effects , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/isolation & purification , Cryoelectron Microscopy , Models, Molecular , Mutation , Phosphoproteins/isolation & purification , RNA Precursors/metabolism , RNA Splicing Factors/isolation & purification , RNA, Messenger/metabolism , RNA-Binding Proteins , Trans-Activators
10.
Bioorg Med Chem Lett ; 28(8): 1336-1341, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29559278

ABSTRACT

The design and synthesis of a novel series of 2,6-disubstituted pyrazine derivatives as CK2 kinase inhibitors is described. Structure-guided optimization of a 5-substituted-3-thiophene carboxylic acid screening hit (3a) led to the development of a lead compound (12b), which shows inhibition in both enzymatic and cellular assays. Subsequent design and hybridization efforts also led to the unexpected identification of analogs with potent PIM kinase activity (14f).


Subject(s)
Casein Kinase II/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Pyrazines/pharmacology , Cell Line, Tumor , Drug Design , Humans , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Pyrazines/chemical synthesis , Pyrazines/chemistry , Pyrazines/pharmacokinetics , Structure-Activity Relationship
11.
J Med Chem ; 59(24): 11079-11097, 2016 12 22.
Article in English | MEDLINE | ID: mdl-28002961

ABSTRACT

Protein lysine methyltransferases (KMTs) have emerged as important regulators of epigenetic signaling. These enzymes catalyze the transfer of donor methyl groups from the cofactor S-adenosylmethionine to specific acceptor lysine residues on histones, leading to changes in chromatin structure and transcriptional regulation. These enzymes also methylate an array of nonhistone proteins, suggesting additional mechanisms by which they influence cellular physiology. SMYD2 is reported to be an oncogenic methyltransferase that represses the functional activity of the tumor suppressor proteins p53 and RB. HTS screening led to identification of five distinct substrate-competitive chemical series. Determination of liganded crystal structures of SMYD2 contributed significantly to "hit-to-lead" design efforts, culminating in the creation of potent and selective inhibitors that were used to understand the functional consequences of SMYD2 inhibition. Taken together, these results have broad implications for inhibitor design against KMTs and clearly demonstrate the potential for developing novel therapies against these enzymes.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , HCT116 Cells , Histone-Lysine N-Methyltransferase/metabolism , Humans , Molecular Structure , Structure-Activity Relationship
12.
ACS Med Chem Lett ; 7(3): 300-5, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26985319

ABSTRACT

The Wnt pathway is an evolutionarily conserved and tightly regulated signaling network with important roles in embryonic development and adult tissue regeneration. Impaired Wnt pathway regulation, arising from mutations in Wnt signaling components, such as Axin, APC, and ß-catenin, results in uncontrolled cell growth and triggers oncogenesis. To explore the reported link between CK2 kinase activity and Wnt pathway signaling, we sought to identify a potent, selective inhibitor of CK2 suitable for proof of concept studies in vivo. Starting from a pyrazolo[1,5-a]pyrimidine lead (2), we identified compound 7h, a potent CK2 inhibitor with picomolar affinity that is highly selectivity against other kinase family enzymes and inhibits Wnt pathway signaling (IC50 = 50 nM) in DLD-1 cells. In addition, compound 7h has physicochemical properties that are suitable for formulation as an intravenous solution, has demonstrated good pharmacokinetics in preclinical species, and exhibits a high level of activity as a monotherapy in HCT-116 and SW-620 xenografts.

13.
Bioorg Med Chem Lett ; 25(24): 5743-7, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26546219

ABSTRACT

The propensity for cancer cells to accumulate additional centrosomes relative to normal cells could be exploited for therapeutic benefit in oncology. Following literature reports that suggested TNKS1 (tankyrase 1) and PARP16 may be involved with spindle structure and function and may play a role in suppressing multi-polar spindle formation in cells with supernumerary centrosomes, we initiated a phenotypic screen to look for small molecule poly (ADP-ribose) polymerase (PARP) enzyme family inhibitors that could produce a multi-polar spindle phenotype via declustering of centrosomes. Screening of AstraZeneca's collection of phthalazinone PARP inhibitors in HeLa cells using high-content screening techniques identified several compounds that produced a multi-polar spindle phenotype at low nanomolar concentrations. Characterization of these compounds across a broad panel of PARP family enzyme assays indicated that they had activity against several PARP family enzymes, including PARP1, 2, 3, 5a, 5b, and 6. Further optimization of these initial hits for improved declustering potency, solubility, permeability, and oral bioavailability resulted in AZ0108, a PARP1, 2, 6 inhibitor that potently inhibits centrosome clustering and is suitable for in vivo efficacy and tolerability studies.


Subject(s)
Centrosome/metabolism , Phthalazines/chemistry , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Administration, Oral , Animals , Binding Sites , Caco-2 Cells , Centrosome/drug effects , Crystallography, X-Ray , Drug Evaluation, Preclinical , HeLa Cells , Humans , Microsomes/metabolism , Molecular Conformation , Molecular Dynamics Simulation , Phthalazines/administration & dosage , Phthalazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Structure, Tertiary , Rats , Tankyrases/antagonists & inhibitors , Tankyrases/metabolism
14.
ACS Chem Biol ; 8(3): 643-50, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23272696

ABSTRACT

Transforming growth factor-ß activated kinase-1 (TAK1) is a member of the mitogen-activated protein kinase kinase kinase (MAP3K) family that regulates several signaling pathways including NF-κB signal transduction and p38 activation. TAK1 deregulation has been implicated in human diseases including cancer and inflammation. Here, we show that, in addition to its kinase activity, TAK1 has intrinsic ATPase activity, that (5Z)-7-Oxozeaenol irreversibly inhibits TAK1, and that sensitivity to (5Z)-7-Oxozeaenol inhibition in hematological cancer cell lines is NRAS mutation status and TAK1 pathway dependent. X-ray crystallographic and mass spectrometric studies showed that (5Z)-7-Oxozeaenol forms a covalent complex with TAK1. Detailed biochemical characterization revealed that (5Z)-7-Oxozeaenol inhibited both the kinase and the ATPase activity of TAK1 following a bi-phase kinetics, consistent with the irreversible inhibition mechanism. In DoHH2 cells, (5Z)-7-Oxozeaenol potently inhibited the p38 phosphorylation driven by TAK1, and the inhibition lasted over 6 h after withdrawal of (5Z)-7-Oxozeaenol. Profiling (5Z)-7-Oxozeaenol in a panel of hematological cancer cells showed that sensitive cell lines tended to carry NRAS mutations and that genes in TAK1 regulated pathways were enriched in sensitive cell lines. Taken together, we have elucidated the molecular mechanism of a TAK1 irreversible inhibitor and laid the foundation for designing next generation TAK1 irreversible inhibitors. The NRAS-TAK1-Wnt signaling network discerned in our study may prove to be useful in patient selection for TAK1 targeted agents in hematological cancers.


Subject(s)
MAP Kinase Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Zearalenone/analogs & derivatives , Cell Line, Tumor , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , MAP Kinase Kinase Kinases/metabolism , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Zearalenone/chemistry , Zearalenone/pharmacology
15.
ACS Med Chem Lett ; 4(8): 800-5, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-24900749

ABSTRACT

In this letter, we describe the design, synthesis, and structure-activity relationship of 5-anilinopyrazolo[1,5-a]pyrimidine inhibitors of CK2 kinase. Property-based optimization of early leads using the 7-oxetan-3-yl amino group led to a series of matched molecular pairs with lower lipophilicity, decreased affinity for human plasma proteins, and reduced binding to the hERG ion channel. Agents in this study were shown to modulate pAKT(S129), a direct substrate of CK2, in vitro and in vivo, and exhibited tumor growth inhibition when administered orally in a murine DLD-1 xenograft.

16.
J Med Chem ; 55(22): 10010-21, 2012 Nov 26.
Article in English | MEDLINE | ID: mdl-23043329

ABSTRACT

Thymidylate kinase (TMK) is an essential enzyme in bacterial DNA synthesis. The deoxythymidine monophosphate (dTMP) substrate binding pocket was targeted in a rational-design, structure-supported effort, yielding a unique series of antibacterial agents showing a novel, induced-fit binding mode. Lead optimization, aided by X-ray crystallography, led to picomolar inhibitors of both Streptococcus pneumoniae and Staphylococcus aureus TMK. MICs < 1 µg/mL were achieved against methicillin-resistant S. aureus (MRSA), S. pneumoniae, and vancomycin-resistant Enterococcus (VRE). Log D adjustments yielded single diastereomers 14 (TK-666) and 46, showing a broad antibacterial spectrum against Gram-positive bacteria and excellent selectivity against the human thymidylate kinase ortholog.


Subject(s)
Anti-Bacterial Agents/pharmacology , Benzoates/pharmacology , Enterococcus/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Nucleoside-Phosphate Kinase/antagonists & inhibitors , Staphylococcus aureus/drug effects , Streptococcus pneumoniae/drug effects , Thymine/analogs & derivatives , Vancomycin Resistance/drug effects , Anti-Bacterial Agents/chemical synthesis , Benzoates/chemical synthesis , Catalytic Domain , Crystallography, X-Ray , Humans , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Nucleoside-Phosphate Kinase/metabolism , Structure-Activity Relationship , Thymine/chemical synthesis , Thymine/pharmacology
17.
Biochem J ; 446(3): 405-13, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22721802

ABSTRACT

GlmU is a bifunctional enzyme with acetyltransferase and uridyltransferase activities, and is essential for the biosynthesis of the bacterial cell wall. Inhibition results in a loss of cell viability. GlmU is therefore considered a potential target for novel antibacterial agents. A HTS (high-throughput screen) identified a series of aminoquinazolines with submicromolar potency against the uridyltransferase reaction. Biochemical and biophysical characterization showed competition with UTP binding. We determined the crystal structure of a representative aminoquinazoline bound to the Haemophilus influenzae isoenzyme at a resolution of 2.0 Å. The inhibitor occupies part of the UTP site, skirts the outer perimeter of the GlcNAc1-P (N-acetylglucosamine-1-phosphate) pocket and anchors a hydrophobic moiety into a lipophilic pocket. Our SAR (structure-activity relationship) analysis shows that all of these interactions are essential for inhibitory activity in this series. The crystal structure suggests that the compound would block binding of UTP and lock GlmU in an apo-enzyme-like conformation, thus interfering with its enzymatic activity. Our lead generation effort provides ample scope for further optimization of these compounds for antibacterial drug discovery.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/chemistry , Acetylglucosamine/analogs & derivatives , Acetylglucosamine/chemistry , Acetylglucosamine/metabolism , Acetyltransferases/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Cell Wall , Crystallography, X-Ray , Haemophilus influenzae/enzymology , Haemophilus influenzae/metabolism , Models, Molecular , Multienzyme Complexes/metabolism , Nucleotidyltransferases/chemistry , Quinazolines/chemistry , Quinazolines/metabolism , Structure-Activity Relationship , Uridine Triphosphate/chemistry , Uridine Triphosphate/metabolism
18.
Bioorg Med Chem Lett ; 22(5): 2063-9, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22305584

ABSTRACT

The design, synthesis and biological evaluation of a series of azabenzimidazole derivatives as TBK1/IKKε kinase inhibitors are described. Starting from a lead compound 1a, iterative design and SAR exploitation of the scaffold led to analogues with nM enzyme potencies against TBK1/IKKε. These compounds also exhibited excellent cellular activity against TBK1. Further structure-based design to improve selectivity over CDK2 and Aurora B resulted in compounds such as 5b-e. These probe compounds will facilitate study of the complex cancer biology of TBK1 and IKKε.


Subject(s)
Benzimidazoles/chemistry , Benzimidazoles/pharmacology , I-kappa B Kinase/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Aurora Kinase B , Aurora Kinases , Aza Compounds/chemistry , Aza Compounds/pharmacology , Cyclin-Dependent Kinase 2/metabolism , Drug Design , HEK293 Cells , Humans , I-kappa B Kinase/metabolism , Models, Molecular , Neoplasms/enzymology , Protein Serine-Threonine Kinases/metabolism , Structure-Activity Relationship
19.
Bioorg Med Chem Lett ; 22(4): 1690-4, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22264476

ABSTRACT

A series of structurally unique Smac mimetics that act as antagonists of inhibitor of apoptosis proteins (IAPs) has been discovered. While most previously described Smac mimetics contain the proline ring (or a similar cyclic motif) found in Smac, a key feature of the compounds described herein is that this ring has been removed. Despite this, compounds in this series potently bind to cIAP1 and elicit the expected phenotype of cIAP1 inhibition in cancer cells. Marked selectivity for cIAP1 over XIAP is observed for these compounds, which is attributed to a slight difference in the binding groove between the two proteins and the resulting steric interactions with the inhibitors. XIAP binding can be improved by constraining the inhibitor so that these unfavorable steric interactions are minimized.


Subject(s)
Amines/chemical synthesis , Drug Design , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/chemistry , Mitochondrial Proteins/chemistry , Piperidines/chemical synthesis , Amines/chemistry , Amines/pharmacology , Apoptosis Regulatory Proteins , Biomimetics , Cell Line, Tumor , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Piperidines/chemistry , Piperidines/pharmacology , Protein Binding/drug effects , Structure-Activity Relationship
20.
ACS Med Chem Lett ; 3(4): 278-83, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-24900464

ABSTRACT

In this paper we describe a series of 3-cyano-5-aryl-7-aminopyrazolo[1,5-a]pyrimidine hits identified by kinase-focused subset screening as starting points for the structure-based design of conformationally constrained 6-acetamido-indole inhibitors of CK2. The synthesis, SAR, and effects of this novel series on Akt signaling and cell proliferation in vitro are described.

SELECTION OF CITATIONS
SEARCH DETAIL
...