Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Med Chem ; 67(6): 4541-4559, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38466661

ABSTRACT

The optimization of an allosteric fragment, discovered by differential scanning fluorimetry, to an in vivo MAT2a tool inhibitor is discussed. The structure-based drug discovery approach, aided by relative binding free energy calculations, resulted in AZ'9567 (21), a potent inhibitor in vitro with excellent preclinical pharmacokinetic properties. This tool showed a selective antiproliferative effect on methylthioadenosine phosphorylase (MTAP) KO cells, both in vitro and in vivo, providing further evidence to support the utility of MAT2a inhibitors as potential anticancer therapies for MTAP-deficient tumors.


Subject(s)
Neoplasms , Humans , Entropy , Methionine Adenosyltransferase/metabolism
2.
J Med Chem ; 67(4): 3090-3111, 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38306388

ABSTRACT

The inhibition of ataxia-telangiectasia mutated (ATM) has been shown to chemo- and radio-sensitize human glioma cells in vitro and therefore might provide an exciting new paradigm in the treatment of glioblastoma multiforme (GBM). The effective treatment of GBM will likely require a compound with the potential to efficiently cross the blood-brain barrier (BBB). Starting from clinical candidate AZD0156, 4, we investigated the imidazoquinolin-2-one scaffold with the goal of improving likely CNS exposure in humans. Strategies aimed at reducing hydrogen bonding, basicity, and flexibility of the molecule were explored alongside modulating lipophilicity. These studies identified compound 24 (AZD1390) as an exceptionally potent and selective inhibitor of ATM with a good preclinical pharmacokinetic profile. 24 showed an absence of human transporter efflux in MDCKII-MDR1-BCRP studies (efflux ratio <2), significant BBB penetrance in nonhuman primate PET studies (Kp,uu 0.33) and was deemed suitable for development as a clinical candidate to explore the radiosensitizing effects of ATM in intracranial malignancies.


Subject(s)
Ataxia Telangiectasia , Glioblastoma , Pyridines , Quinolones , Animals , Humans , Blood-Brain Barrier/metabolism , Ataxia Telangiectasia/drug therapy , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Ataxia Telangiectasia Mutated Proteins , Neoplasm Proteins , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Glioblastoma/drug therapy
3.
J Med Chem ; 67(2): 1500-1512, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38227216

ABSTRACT

Casitas B-lymphoma proto-oncogene-b (Cbl-b), a member of the Cbl family of RING finger E3 ubiquitin ligases, has been demonstrated to play a central role in regulating effector T-cell function. Multiple studies using gene-targeting approaches have provided direct evidence that Cbl-b negatively regulates T, B, and NK cell activation via a ubiquitin-mediated protein modulation. Thus, inhibition of Cbl-b ligase activity can lead to immune activation and has therapeutic potential in immuno-oncology. Herein, we describe the discovery and optimization of an arylpyridone series as Cbl-b inhibitors by structure-based drug discovery to afford compound 31. This compound binds to Cbl-b with an IC50 value of 30 nM and induces IL-2 production in T-cells with an EC50 value of 230 nM. Compound 31 also shows robust intracellular target engagement demonstrated through inhibition of Cbl-b autoubiquitination, inhibition of ubiquitin transfer to ZAP70, and the cellular modulation of phosphorylation of a downstream signal within the TCR axis.


Subject(s)
Proto-Oncogene Proteins c-cbl , Ubiquitin-Protein Ligases , Proto-Oncogene Proteins c-cbl/metabolism , Ubiquitin-Protein Ligases/metabolism , T-Lymphocytes/metabolism , Phosphorylation , Ubiquitin/metabolism
4.
J Med Chem ; 66(13): 9147-9160, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37395055

ABSTRACT

The glycine to cysteine mutation at codon 12 of Kirsten rat sarcoma (KRAS) represents an Achilles heel that has now rendered this important GTPase druggable. Herein, we report our structure-based drug design approach that led to the identification of 14, AZD4747, a clinical development candidate for the treatment of KRASG12C-positive tumors, including the treatment of central nervous system (CNS) metastases. Building on our earlier discovery of C5-tethered quinazoline AZD4625, excision of a usually critical pyrimidine ring yielded a weak but brain-penetrant start point which was optimized for potency and DMPK. Key design principles and measured parameters that give high confidence in CNS exposure are discussed. During optimization, divergence between rodent and non-rodent species was observed in CNS exposure, with primate PET studies ultimately giving high confidence in the expected translation to patients. AZD4747 is a highly potent and selective inhibitor of KRASG12C with an anticipated low clearance and high oral bioavailability profile in humans.


Subject(s)
Antineoplastic Agents , Lung Neoplasms , Neoplasms , Animals , Humans , Antineoplastic Agents/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Neoplasms/drug therapy , Drug Design , Glycine/therapeutic use , Mutation , Lung Neoplasms/drug therapy
5.
Bioorg Med Chem ; 63: 116729, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35439688

ABSTRACT

In this article, we report the discovery of a series of pyrimidopyridones as inhibitors of IRAK4 kinase. From a previously disclosed 5-azaquinazoline series, we found that switching the pyridine ring for an N-substituted pyridone gave a novel hinge binding scaffold which retained potency against IRAK4. Importantly, introduction of the carbonyl established an internal hydrogen bond with the 4-NH, establishing a conformational lock and allowing truncation of the large basic substituent to a 1-methylcyclopyl group. Subsequent optimisation, facilitated by X-ray crystal structures, allowed identification of preferred substituents at both the pyridone core and pyrazole. Subsequent combinations of optimal groups allowed control of lipophilicity and identification of potent and selective inhibitors of IRAK4 with better in vitro permeability and lower clearance.


Subject(s)
Interleukin-1 Receptor-Associated Kinases , Pyridones , Molecular Conformation , Pyridones/pharmacology , Structure-Activity Relationship
7.
Bioorg Med Chem ; 28(23): 115815, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33091850

ABSTRACT

In this article, we report our efforts towards improving in vitro human clearance in a series of 5-azaquinazolines through a series of C4 truncations and C2 expansions. Extensive DMPK studies enabled us to tackle high Aldehyde Oxidase (AO) metabolism and unexpected discrepancies in human hepatocyte and liver microsomal intrinsic clearance. Our efforts culminated with the discovery of 5-azaquinazoline 35, which also displayed exquisite selectivity for IRAK4, and showed synergistic in vitro activity against MyD88/CD79 double mutant ABC-DLBCL in combination with the covalent BTK inhibitor acalabrutinib.


Subject(s)
Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/metabolism , Quinazolines/chemistry , Aldehyde Oxidase/metabolism , Animals , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Dogs , Drug Stability , Half-Life , Hepatocytes/metabolism , Humans , Interleukin-1 Receptor-Associated Kinases/metabolism , Mice , Microsomes, Liver/metabolism , Molecular Dynamics Simulation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Quinazolines/metabolism , Quinazolines/pharmacology , Rats , Structure-Activity Relationship
8.
J Med Chem ; 62(21): 9918-9930, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31622099

ABSTRACT

In this article, we report the discovery of a series of 5-azaquinazolines as selective IRAK4 inhibitors. From modestly potent quinazoline 4, we introduced a 5-aza substitution to mask the 4-NH hydrogen bond donor (HBD). This allowed us to substitute the core with a 2-aminopyrazole, which showed large gains in cellular potency despite the additional formal HBD. Further optimization led to 6-cyanomethyl-5-azaquinazoline 13, a selective IRAK4 inhibitor, which proved efficacious in combination with ibrutinib, while showing very little activity as a single agent up to 100 mg/kg. This contrasted to previously reported IRAK4 inhibitors that exhibited efficacy in the same model as single agents and was attributed to the enhanced specificity of 13 toward IRAK4.


Subject(s)
Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Lymphoma, Large B-Cell, Diffuse/drug therapy , Molecular Targeted Therapy , Myeloid Differentiation Factor 88/genetics , Quinazolines/chemistry , Quinazolines/pharmacology , Administration, Oral , Animals , Cell Line, Tumor , Drug Design , Female , Humans , Interleukin-1 Receptor-Associated Kinases/chemistry , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Models, Molecular , Mutation , Protein Conformation , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Quinazolines/administration & dosage , Quinazolines/pharmacokinetics , Rats , Rats, Wistar , Structure-Activity Relationship , Tissue Distribution , Xenograft Model Antitumor Assays
9.
J Med Chem ; 62(21): 9418-9437, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31361481

ABSTRACT

The three-dimensional conformations adopted by a free ligand in solution impact bioactivity and physicochemical properties. Solution 1D NMR spectra inherently contain information on ligand conformational flexibility and three-dimensional shape, as well as the propensity of the free ligand to fully preorganize into the bioactive conformation. Herein we discuss some key learnings, distilled from our experience developing potent and selective synthetic macrocyclic inhibitors, including Mcl-1 clinical candidate AZD5991. Case studies have been selected from recent oncology research projects, demonstrating how 1D NMR conformational signatures can complement X-ray protein-ligand structural information to guide medicinal chemistry optimization. Learning to extract free ligand conformational information from routinely available 1D NMR signatures has proven to be fast enough to guide medicinal chemistry decisions within design cycles for compound optimization.


Subject(s)
Drug Design , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Kinetics , Ligands , Macrocyclic Compounds/chemical synthesis , Magnetic Resonance Spectroscopy , Models, Molecular , Myeloid Cell Leukemia Sequence 1 Protein/chemistry , Protein Conformation , Structure-Activity Relationship
10.
ACS Chem Biol ; 13(11): 3131-3141, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30335946

ABSTRACT

B-cell lymphoma 6 (BCL6) inhibition is a promising mechanism for treating hematological cancers but high quality chemical probes are necessary to evaluate its therapeutic potential. Here we report potent BCL6 inhibitors that demonstrate cellular target engagement and exhibit exquisite selectivity for BCL6 based on mass spectrometry analyses following chemical proteomic pull down. Importantly, a proteolysis-targeting chimera (PROTAC) was also developed and shown to significantly degrade BCL6 in a number of diffuse large B-cell lymphoma (DLBCL) cell lines, but neither BCL6 inhibition nor degradation selectively induced marked phenotypic response. To investigate, we monitored PROTAC directed BCL6 degradation in DLBCL OCI-Ly1 cells by immunofluorescence and discovered a residual BCL6 population. Analysis of subcellular fractions also showed incomplete BCL6 degradation in all fractions despite having measurable PROTAC concentrations, together providing a rationale for the weak antiproliferative response seen with both BCL6 inhibitor and degrader. In summary, we have developed potent and selective BCL6 inhibitors and a BCL6 PROTAC that effectively degraded BCL6, but both modalities failed to induce a significant phenotypic response in DLBCL despite achieving cellular concentrations.


Subject(s)
Antineoplastic Agents/pharmacology , Proto-Oncogene Proteins c-bcl-6/antagonists & inhibitors , Quinolones/pharmacology , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Adaptor Proteins, Signal Transducing , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Cell Line, Tumor , HEK293 Cells , Humans , Ligands , Lymphoma, Large B-Cell, Diffuse/drug therapy , Peptide Hydrolases/metabolism , Protein Binding , Proteolysis , Proto-Oncogene Proteins c-bcl-6/chemistry , Proto-Oncogene Proteins c-bcl-6/metabolism , Quinolones/chemical synthesis , Quinolones/metabolism , Thalidomide/chemical synthesis , Thalidomide/metabolism , Ubiquitin-Protein Ligases/metabolism
11.
Bioorg Med Chem ; 26(4): 913-924, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29398441

ABSTRACT

We have developed a series of orally efficacious IRAK4 inhibitors, based on a scaffold hopping strategy and using rational structure based design. Efforts to tackle low permeability and high efflux in our previously reported pyrrolopyrimidine series (Scott et al., 2017) led to the identification of pyrrolotriazines which contained one less formal hydrogen bond donor and were intrinsically more lipophilic. Further optimisation of substituents on this pyrrolotriazine core culminated with the discovery of 30 as a promising in vivo probe to assess the potential of IRAK4 inhibition for the treatment of MyD88 mutant DLBCL in combination with a BTK inhibitor. When tested in an ABC-DLBCL model with a dual MyD88/CD79 mutation (OCI-LY10), 30 demonstrated tumour regressions in combination with ibrutinib.


Subject(s)
Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Pyrroles/chemistry , Thiazines/chemistry , Animals , Binding Sites , Caco-2 Cells , Dogs , Drug Design , Half-Life , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Interleukin-1 Receptor-Associated Kinases/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Molecular Dynamics Simulation , Mutation , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Permeability/drug effects , Protein Kinases/chemistry , Protein Kinases/metabolism , Pyrroles/pharmacokinetics , Pyrroles/pharmacology , Rats , Structure-Activity Relationship , Thiazines/pharmacokinetics , Thiazines/pharmacology
12.
J Med Chem ; 60(24): 10071-10091, 2017 12 28.
Article in English | MEDLINE | ID: mdl-29172502

ABSTRACT

Herein we report the optimization of a series of pyrrolopyrimidine inhibitors of interleukin-1 receptor associated kinase 4 (IRAK4) using X-ray crystal structures and structure based design to identify and optimize our scaffold. Compound 28 demonstrated a favorable physicochemical and kinase selectivity profile and was identified as a promising in vivo tool with which to explore the role of IRAK4 inhibition in the treatment of mutant MYD88L265P diffuse large B-cell lymphoma (DLBCL). Compound 28 was shown to be capable of demonstrating inhibition of NF-κB activation and growth of the ABC subtype of DLBCL cell lines in vitro at high concentrations but showed greater effects in combination with a BTK inhibitor at lower concentrations. In vivo, the combination of compound 28 and ibrutinib led to tumor regression in an ABC-DLBCL mouse model.


Subject(s)
Antineoplastic Agents/pharmacology , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Lymphoma, Large B-Cell, Diffuse/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Crystallography, X-Ray , Dogs , Female , Humans , Interleukin-1 Receptor-Associated Kinases/chemistry , Lymphoma, Large B-Cell, Diffuse/genetics , Magnetic Resonance Spectroscopy , Male , Mice, SCID , Mutation , Myeloid Differentiation Factor 88/genetics , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/chemistry , Pyrroles/chemistry , Rats, Wistar , Structure-Activity Relationship , Xenograft Model Antitumor Assays
14.
J Med Chem ; 60(10): 4386-4402, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28485934

ABSTRACT

Inhibition of the protein-protein interaction between B-cell lymphoma 6 (BCL6) and corepressors has been implicated as a therapeutic target in diffuse large B-cell lymphoma (DLBCL) cancers and profiling of potent and selective BCL6 inhibitors are critical to test this hypothesis. We identified a pyrazolo[1,5-a]pyrimidine series of BCL6 binders from a fragment screen in parallel with a virtual screen. Using structure-based drug design, binding affinity was increased 100000-fold. This involved displacing crystallographic water, forming new ligand-protein interactions and a macrocyclization to favor the bioactive conformation of the ligands. Optimization for slow off-rate constant kinetics was conducted as well as improving selectivity against an off-target kinase, CK2. Potency in a cellular BCL6 assay was further optimized to afford highly selective probe molecules. Only weak antiproliferative effects were observed across a number of DLBCL lines and a multiple myeloma cell line without a clear relationship to BCL6 potency. As a result, we conclude that the BCL6 hypothesis in DLBCL cancer remains unproven.


Subject(s)
Protein Interaction Maps/drug effects , Proto-Oncogene Proteins c-bcl-6/metabolism , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Molecular Docking Simulation , Proto-Oncogene Proteins c-bcl-6/antagonists & inhibitors
15.
Nat Chem Biol ; 12(12): 1097-1104, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27775716

ABSTRACT

Proteins of the bromodomain and extraterminal (BET) family, in particular bromodomain-containing protein 4 (BRD4), are of great interest as biological targets. BET proteins contain two separate bromodomains, and existing inhibitors bind to them monovalently. Here we describe the discovery and characterization of probe compound biBET, capable of engaging both bromodomains simultaneously in a bivalent, in cis binding mode. The evidence provided here was obtained in a variety of biophysical and cellular experiments. The bivalent binding results in very high cellular potency for BRD4 binding and pharmacological responses such as disruption of BRD4-mediator complex subunit 1 foci with an EC50 of 100 pM. These compounds will be of considerable utility as BET/BRD4 chemical probes. This work illustrates a novel concept in ligand design-simultaneous targeting of two separate domains with a drug-like small molecule-providing precedent for a potentially more effective paradigm for developing ligands for other multi-domain proteins.


Subject(s)
Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/chemistry , Protein Domains/drug effects , Small Molecule Libraries/pharmacology , Transcription Factors/antagonists & inhibitors , Transcription Factors/chemistry , Apoptosis/drug effects , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Ligands , Models, Molecular , Molecular Structure , Nuclear Proteins/metabolism , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Substrate Specificity , Transcription Factors/metabolism
16.
Bioorg Med Chem Lett ; 25(16): 3189-93, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26087940

ABSTRACT

The discovery and optimisation of novel, potent and selective small molecule inhibitors of the α-isoform of type III phosphatidylinositol-4-kinase (PI4Kα) are described. Lead compounds show cellular activity consistent with their PI4Kα potency inhibiting the accumulation of IP1 after PDGF stimulation and reducing cellular PIP, PIP2 and PIP3 levels. Hence, these compounds are useful in vitro tools to delineate the complex biological pathways involved in signalling through PI4Kα.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Drug Design , High-Throughput Screening Assays , Humans , Inositol Phosphates/metabolism , Minor Histocompatibility Antigens , Models, Molecular , Platelet-Derived Growth Factor/pharmacology , Signal Transduction/drug effects , Small Molecule Libraries , Structure-Activity Relationship
17.
ACS Med Chem Lett ; 6(3): 254-9, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25815142

ABSTRACT

The canonical Wnt pathway plays an important role in embryonic development, adult tissue homeostasis, and cancer. Germline mutations of several Wnt pathway components, such as Axin, APC, and ß-catenin, can lead to oncogenesis. Inhibition of the poly(ADP-ribose) polymerase (PARP) catalytic domain of the tankyrases (TNKS1 and TNKS2) is known to inhibit the Wnt pathway via increased stabilization of Axin. In order to explore the consequences of tankyrase and Wnt pathway inhibition in preclinical models of cancer and its impact on normal tissue, we sought a small molecule inhibitor of TNKS1/2 with suitable physicochemical properties and pharmacokinetics for hypothesis testing in vivo. Starting from a 2-phenyl quinazolinone hit (compound 1), we discovered the pyrrolopyrimidinone compound 25 (AZ6102), which is a potent TNKS1/2 inhibitor that has 100-fold selectivity against other PARP family enzymes and shows 5 nM Wnt pathway inhibition in DLD-1 cells. Moreover, compound 25 can be formulated well in a clinically relevant intravenous solution at 20 mg/mL, has demonstrated good pharmacokinetics in preclinical species, and shows low Caco2 efflux to avoid possible tumor resistance mechanisms.

18.
J Med Chem ; 57(14): 6128-40, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-24967667

ABSTRACT

Ghrelin plays a major physiological role in the control of food intake, and inverse agonists of the ghrelin receptor (GHS-R1a) are widely considered to offer utility as antiobesity agents by lowering the set-point for hunger between meals. We identified an acylurea series of ghrelin modulators from high throughput screening and optimized binding affinity through structure-activity relationship studies. Furthermore, we identified specific substructural changes, which switched partial agonist activity to inverse agonist activity, and optimized physicochemical and DMPK properties to afford the non-CNS penetrant inverse agonist 22 (AZ-GHS-22) and the CNS penetrant inverse agonist 38 (AZ-GHS-38). Free feeding efficacy experiments showed that CNS exposure was necessary to obtain reduced food intake in mice, and it was demonstrated using GHS-R1a null and wild-type mice that this effect operates through a mechanism involving GHS-R1a.


Subject(s)
Drug Inverse Agonism , Receptors, Ghrelin/agonists , Receptors, Ghrelin/antagonists & inhibitors , Urea/analogs & derivatives , Urea/pharmacology , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Receptors, Ghrelin/metabolism , Structure-Activity Relationship , Urea/chemistry
19.
J Med Chem ; 57(3): 970-86, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24422550

ABSTRACT

11ß-Hydroxysteroid dehydrogenase type 1 (11ß-HSD1) has been widely considered by the pharmaceutical industry as a target to treat metabolic syndrome in type II diabetics. We hypothesized that central nervous system (CNS) penetration might be required to see efficacy. Starting from a previously reported pyrimidine compound, we removed hydrogen-bond donors to yield 3, which had modest CNS penetration. More significant progress was achieved by changing the core to give 40, which combines good potency and CNS penetration. Compound 40 was dosed to diet-induced obese (DIO) mice and gave excellent target engagement in the liver and high free exposures of drug, both peripherally and in the CNS. However, no body weight reduction or effects on glucose or insulin were observed in this model. Similar data were obtained with a structurally diverse thiazole compound 51. This work casts doubt on the hypothesis that localized tissue modulation of 11ß-HSD1 activity alleviates metabolic syndrome.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Adamantane/analogs & derivatives , Adamantane/chemical synthesis , Brain/metabolism , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/chemical synthesis , Metabolic Syndrome/drug therapy , 11-beta-Hydroxysteroid Dehydrogenase Type 1/chemistry , Adamantane/pharmacokinetics , Adamantane/pharmacology , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Crystallography, X-Ray , Cyclopropanes/chemical synthesis , Cyclopropanes/pharmacokinetics , Cyclopropanes/pharmacology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Dietary Fats/administration & dosage , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Insulin/blood , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Liver/drug effects , Liver/metabolism , Male , Metabolic Syndrome/metabolism , Metabolic Syndrome/physiopathology , Mice , Mice, Inbred C57BL , Mice, Obese , Models, Molecular , Pyrazoles/chemical synthesis , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/pharmacokinetics , Thiazoles/pharmacology
20.
Chem Commun (Camb) ; 50(40): 5388-90, 2014 May 25.
Article in English | MEDLINE | ID: mdl-24366037

ABSTRACT

Two series of inhibitors of type III phosphatidylinositol-4-kinase were identified by high throughput screening and optimised to derive probe compounds that independently and selectively inhibit the α- and the ß-isoforms with no significant activity towards related kinases in the pathway. In a cellular environment, inhibition of the α- but not the ß-subtype led to a reduction in phosphatidylinositol-4-phosphate and phosphatidylinositol-4,5-bisphosphate concentration, causing inhibition of inositol-1-phosphate formation and inhibition of proliferation in a panel of cancer cell lines.


Subject(s)
1-Phosphatidylinositol 4-Kinase/antagonists & inhibitors , Inositol Phosphates/antagonists & inhibitors , Neoplasms/drug therapy , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol Phosphates/metabolism , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Cell Proliferation/drug effects , High-Throughput Screening Assays , Humans , Inositol Phosphates/metabolism , Models, Molecular , Molecular Structure , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction/drug effects , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...