Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Antimicrob Agents Chemother ; 68(3): e0106423, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38349161

ABSTRACT

Screening a library of 1,200 preselected kinase inhibitors for anti-human rhinovirus 2 (HRV-2) activity in HeLa cells identified a class of epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKI) as effective virus blockers. These were based on the 4-anilinoquinazoline-7-oxypiperidine scaffold, with the most potent representative AZ5385 inhibiting the virus with EC50 of 0.35 µM. Several structurally related analogs confirmed activity in the low µM range, while interestingly, other TKIs targeting EGFR lacked anti-HRV-2 activity. To further probe this lack of association between antiviral activity and EGFR inhibition, we stained infected cells with antibodies specific for activated EGFR (Y1068) and did not observe a dependency on EGFR-TK activity. Instead, consecutive passages of HRV-2 in HeLa cells in the presence of a compound and subsequent nucleotide sequence analysis of resistant viral variants identified the S181T and T210A alterations in the major capsid VP1 protein, with both residues located in the vicinity of a known hydrophobic pocket on the viral capsid. Further characterization of the antiviral effects of AZ5385 showed a modest virus-inactivating (virucidal) activity, while anti-HRV-2 activity was still evident when the inhibitor was added as late as 10 h post infection. The RNA copy/infectivity ratio of HRV-2 propagated in AZ5385 presence was substantially higher than that of control HRV indicating that the compound preferentially targeted HRV progeny virions during their maturation in infected cells. Besides HRV, the compound showed anti-respiratory syncytial virus activity, which warrants its further studies as a candidate compound against viral respiratory infections.


Subject(s)
Rhinovirus , Humans , Rhinovirus/chemistry , Rhinovirus/genetics , HeLa Cells , Capsid Proteins , Antiviral Agents/chemistry , ErbB Receptors
2.
ACS Med Chem Lett ; 14(12): 1760-1766, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38116421

ABSTRACT

A high-throughput fragment-based screen has been employed to discover a series of quinazolinone inositol hexakisphosphate kinase (IP6K) inhibitors. IP6Ks have been studied for their role in glucose homeostasis, metabolic disease, fatty liver disease, chronic kidney disease, blood coagulation, neurological development, and psychiatric disease. IP6Ks phosphorylate inositol hexakisphosphate (IP6) to form pyrophosphate 5-diphospho-1,2,3,4,6-pentakisphosphate (IP7). Molecular docking studies and investigation of structure-activity relationships around the quinazolinone core resulted in compounds with submicromolar potency and interesting selectivity for IP6K1 versus the closely related IP6K2 and IP6K3 isoforms.

3.
Nature ; 618(7963): 159-168, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37225977

ABSTRACT

Harnessing the potential beneficial effects of kinase signalling through the generation of direct kinase activators remains an underexplored area of drug development1-5. This also applies to the PI3K signalling pathway, which has been extensively targeted by inhibitors for conditions with PI3K overactivation, such as cancer and immune dysregulation. Here we report the discovery of UCL-TRO-1938 (referred to as 1938 hereon), a small-molecule activator of the PI3Kα isoform, a crucial effector of growth factor signalling. 1938 allosterically activates PI3Kα through a distinct mechanism by enhancing multiple steps of the PI3Kα catalytic cycle and causes both local and global conformational changes in the PI3Kα structure. This compound is selective for PI3Kα over other PI3K isoforms and multiple protein and lipid kinases. It transiently activates PI3K signalling in all rodent and human cells tested, resulting in cellular responses such as proliferation and neurite outgrowth. In rodent models, acute treatment with 1938 provides cardioprotection from ischaemia-reperfusion injury and, after local administration, enhances nerve regeneration following nerve crush. This study identifies a chemical tool to directly probe the PI3Kα signalling pathway and a new approach to modulate PI3K activity, widening the therapeutic potential of targeting these enzymes through short-term activation for tissue protection and regeneration. Our findings illustrate the potential of activating kinases for therapeutic benefit, a currently largely untapped area of drug development.


Subject(s)
Nerve Regeneration , Humans , Neoplasms/drug therapy , Nerve Regeneration/drug effects , Protein Isoforms/agonists , Signal Transduction/drug effects , Class I Phosphatidylinositol 3-Kinases/chemistry , Class I Phosphatidylinositol 3-Kinases/drug effects , Cardiotonic Agents/pharmacology , Animals , Biocatalysis/drug effects , Protein Conformation/drug effects , Neurites/drug effects , Reperfusion Injury/prevention & control , Nerve Crush , Cell Proliferation/drug effects
4.
Stem Cell Reports ; 18(2): 555-569, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36669494

ABSTRACT

Marfan syndrome (MFS) is a rare connective tissue disorder caused by mutations in FBN1. Patients with MFS notably suffer from aortic aneurysm and dissection. Despite considerable effort, animal models have proven to be poorly predictive for therapeutic intervention in human aortic disease. Patient-derived induced pluripotent stem cells can be differentiated into vascular smooth muscle cells (VSMCs) and recapitulate major features of MFS. We have screened 1,022 small molecules in our in vitro model, exploiting the highly proteolytic nature of MFS VSMCs, and identified 36 effective compounds. Further analysis identified GSK3ß as a recurring target in the compound screen. GSK3ß inhibition/knockdown did not ameliorate the proliferation defect in MFS-VSMCs but improved MFS-VSMC proteolysis and apoptosis and partially rescued fibrillin-1 deposition. To conclude, we have identified GSK3ß as a novel target for MFS, forming the foundation for future work in MFS and other aortic diseases.


Subject(s)
Induced Pluripotent Stem Cells , Marfan Syndrome , Animals , Humans , Marfan Syndrome/genetics , Muscle, Smooth, Vascular , Aorta , Glycogen Synthase Kinase 3 beta
5.
Int J Mol Sci ; 21(24)2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33334026

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) has a large impact on global health. At the onset of disease, NAFLD is characterized by hepatic steatosis defined by the accumulation of triglycerides stored as lipid droplets. Developing therapeutics against NAFLD and progression to non-alcoholic steatohepatitis (NASH) remains a high priority in the medical and scientific community. Drug discovery programs to identify potential therapeutic compounds have supported high throughput/high-content screening of in vitro human-relevant models of NAFLD to accelerate development of efficacious anti-steatotic medicines. Human induced pluripotent stem cell (hiPSC) technology is a powerful platform for disease modeling and therapeutic assessment for cell-based therapy and personalized medicine. In this study, we applied AstraZeneca's chemogenomic library, hiPSC technology and multiplexed high content screening to identify compounds that significantly reduced intracellular neutral lipid content. Among 13,000 compounds screened, we identified hits that protect against hiPSC-derived hepatic endoplasmic reticulum stress-induced steatosis by a mechanism of action including inhibition of the cyclin D3-cyclin-dependent kinase 2-4 (CDK2-4)/CCAAT-enhancer-binding proteins (C/EBPα)/diacylglycerol acyltransferase 2 (DGAT2) pathway, followed by alteration of the expression of downstream genes related to NAFLD. These findings demonstrate that our phenotypic platform provides a reliable approach in drug discovery, to identify novel drugs for treatment of fatty liver disease as well as to elucidate their underlying mechanisms.


Subject(s)
Drug Screening Assays, Antitumor , Endoplasmic Reticulum Stress/drug effects , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Induced Pluripotent Stem Cells/cytology , Lipid Metabolism/drug effects , Signal Transduction/drug effects , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , Computational Biology/methods , Cyclin-Dependent Kinase 2/metabolism , Diacylglycerol O-Acyltransferase/metabolism , Drug Screening Assays, Antitumor/methods , High-Throughput Nucleotide Sequencing , Humans , Lipid Droplets/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Protein Kinase Inhibitors/pharmacology
6.
Biochem J ; 477(17): 3329-3347, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32815546

ABSTRACT

Despite being catalytically defective, pseudokinases are typically essential players of cellular signalling, acting as allosteric regulators of their active counterparts. Deregulation of a growing number of pseudokinases has been linked to human diseases, making pseudokinases therapeutic targets of interest. Pseudokinases can be dynamic, adopting specific conformations critical for their allosteric function. Interfering with their allosteric role, with small molecules that would lock pseudokinases in a conformation preventing their productive partner interactions, is an attractive therapeutic strategy to explore. As a well-known allosteric activator of epidermal growth factor receptor family members, and playing a major part in cancer progression, the pseudokinase HER3 is a relevant context in which to address the potential of pseudokinases as drug targets for the development of allosteric inhibitors. In this proof-of-concept study, we developed a multiplex, medium-throughput thermal shift assay screening strategy to assess over 100 000 compounds and identify selective small molecule inhibitors that would trap HER3 in a conformation which is unfavourable for the formation of an active HER2-HER3 heterodimer. As a proof-of-concept compound, AC3573 bound with some specificity to HER3 and abrogated HER2-HER3 complex formation and downstream signalling in cells. Our study highlights the opportunity to identify new molecular mechanisms of action interfering with the biological function of pseudokinases.


Subject(s)
Protein Kinase Inhibitors , Receptor, ErbB-3 , Allosteric Regulation , Animals , CHO Cells , Cricetulus , Drug Screening Assays, Antitumor , Humans , Proof of Concept Study , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/antagonists & inhibitors , Receptor, ErbB-3/chemistry , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism
7.
Cell Death Discov ; 6: 6, 2020.
Article in English | MEDLINE | ID: mdl-32123582

ABSTRACT

Regulated necrosis or necroptosis, mediated by receptor-interacting kinase 1 (RIPK1), RIPK3 and pseudokinase mixed lineage kinase domain-like protein (MLKL), contributes to the pathogenesis of inflammatory, infectious and degenerative diseases. Recently identified necroptosis inhibitors display moderate specificity, suboptimal pharmacokinetics, off-target effects and toxicity, preventing these molecules from reaching the clinic. Here, we developed a cell-based high-throughput screening (HTS) cascade for the identification of small-molecule inhibitors of necroptosis. From the initial library of over 250,000 compounds, the primary screening phase identified 356 compounds that strongly inhibited TNF-α-induced necroptosis, but not apoptosis, in human and murine cell systems, with EC50 < 6.7 µM. From these, 251 compounds were tested for RIPK1 and/or RIPK3 kinase inhibitory activity; some were active and several have novel mechanisms of action. Based on specific chemical descriptors, 110 compounds proceeded into the secondary screening cascade, which then identified seven compounds with maximum ability to reduce MLKL activation, IC50 >100 µM, EC50 2.5-11.5 µM under long-term necroptosis execution in murine fibroblast L929 cells, and full protection from ATP depletion and membrane leakage in human and murine cells. As a proof of concept, compound SN-6109, with binding mode to RIPK1 similar to that of necrostatin-1, confirmed RIPK1 inhibitory activity and appropriate pharmacokinetic properties. SN-6109 was further tested in mice, showing efficacy against TNF-α-induced systemic inflammatory response syndrome. In conclusion, a phenotypic-driven HTS cascade promptly identified robust necroptosis inhibitors with in vivo activity, currently undergoing further medicinal chemistry optimization. Notably, the novel hits highlight the opportunity to identify new molecular mechanisms of action in necroptosis.

8.
Bioorg Med Chem Lett ; 27(13): 3030-3035, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28526367

ABSTRACT

We report the discovery of a novel aminopyrazine series of PI3Kα inhibitors, designed by hybridizing two known scaffolds of PI3K inhibitors. We describe the progress achieved from the first compounds plagued with poor general kinase selectivity to compounds showing high selectivity for PI3Kα over PI3Kß and excellent general kinase selectivity. This effort culminated with the identification of compound 5 displaying high potency and selectivity, and suitable physiochemical and pharmacokinetic properties for oral administration. In vivo, compound 5 showed good inhibition of tumour growth (86% tumour growth inhibition at 50mg/kg twice daily orally) in the MCF7 xenograft model in mice.


Subject(s)
Drug Discovery , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , Class I Phosphatidylinositol 3-Kinases , Dose-Response Relationship, Drug , Humans , Molecular Docking Simulation , Molecular Structure , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrazines/chemical synthesis , Pyrazines/chemistry , Structure-Activity Relationship
9.
Bioorg Med Chem Lett ; 27(9): 1949-1954, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28347666

ABSTRACT

Attempts to lock the active conformation of compound 4, a PI3Kß/δ inhibitor (PI3Kß cell IC50 0.015µM), led to the discovery of a series of 8-(1-phenylpyrrolidin-2-yl)-6-carboxamide-2-morpholino-4H-chromen-4-ones, which showed high levels of potency and selectivity as PI3Kß/δ inhibitors. Compound 10 proved exquisitely potent and selective: PI3Kß cell IC50 0.0011µM in PTEN null MDA-MB-468 cell and PI3Kδ cell IC50 0.014µM in Jeko-1 B-cell, and exhibited suitable physical properties for oral administration. In vivo, compound 10 showed profound pharmacodynamic modulation of AKT phosphorylation in a mouse PTEN-null PC3 prostate tumour xenograft after a single oral dose and gave excellent tumour growth inhibition in the same model after chronic oral dosing. Based on these results, compound 10 was selected as one of our PI3Kß/δ preclinical candidates.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Benzopyrans/chemistry , Benzopyrans/therapeutic use , PTEN Phosphohydrolase/genetics , Phosphoinositide-3 Kinase Inhibitors , Prostatic Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Benzopyrans/pharmacokinetics , Benzopyrans/pharmacology , Cell Line, Tumor , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Dogs , Gene Deletion , Humans , Male , Mice, Nude , Molecular Docking Simulation , Morpholinos/chemistry , Morpholinos/pharmacokinetics , Morpholinos/pharmacology , Morpholinos/therapeutic use , Prostate/drug effects , Prostate/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
10.
Bioorg Med Chem Lett ; 26(9): 2318-23, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26996374

ABSTRACT

We report the discovery and optimisation of a series of 8-(2,3-dihydro-1,4-benzoxazin-4-ylmethyl)-2-morpholino-4-oxo-chromene-6-carboxamides, leading to compound 16 as a potent and selective PI3Kß/δ inhibitor: PI3Kß cell IC50 0.012 µM (in PTEN null MDA-MB-468 cell) and PI3Kδ cell IC50 0.047 µM (in Jeko-1 B-cell), with good pharmacokinetics and physical properties. In vivo, 16 showed profound pharmacodynamic modulation of AKT phosphorylation in a mouse PTEN-deficient PC3 prostate tumour xenograft after a single oral dose and gave excellent tumour growth inhibition in the same model after chronic oral dosing. Compound 16 was selected as a preclinical candidate for the treatment of PTEN-deficient tumours.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Morpholinos/chemistry , Morpholinos/pharmacology , PTEN Phosphohydrolase/genetics , Phosphoinositide-3 Kinase Inhibitors , Animals , Cell Line, Tumor , Humans , Mice , Phosphorylation
11.
Bioorg Med Chem Lett ; 25(22): 5155-62, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26475521

ABSTRACT

Starting from potent inhibitors of PI3Kα having poor general kinase selectivity (e.g., 1 and 2), optimisation of this series led to the identification of 25, a potent inhibitor of PI3Kα (wild type, E545K and H1047R mutations) and PI3Kδ, selective versus PI3Kß and PI3Kγ, with excellent general kinase selectivity. Compound 25 displayed low metabolic turnover and suitable physical properties for oral administration. In vivo, compound 25 showed pharmacodynamic modulation of AKT phosphorylation and near complete inhibition of tumour growth (93% tumour growth inhibition) in a murine H1047R PI3Kα mutated SKOV-3 xenograft tumour model after chronic oral administration at 25mg/kg b.i.d. Compound 25, also known as AZD8835, is currently in phase I clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Oxadiazoles/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Dogs , Humans , Mice , Mice, Nude , Mice, SCID , Molecular Docking Simulation , Oxadiazoles/chemical synthesis , Piperidines/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Rats , Xenograft Model Antitumor Assays
12.
Bioorg Med Chem Lett ; 25(13): 2679-85, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25980912

ABSTRACT

Starting from compound 1, a potent PI3Kα inhibitor having poor general kinase selectivity, we used structural data and modelling to identify key exploitable differences between PI3Kα and the other kinases. This approach led us to design chemical modifications of the central pyrazole, which solved the poor kinase selectivity seen as a strong liability for the initial compound 1. Amongst the modifications explored, a 1,3,4-triazole ring (as in compound 4) as a replacement of the initial pyrazole provided good potency against PI3Kα, with excellent kinase selectivity.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Amino Acid Sequence , Binding Sites , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/genetics , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Drug Design , Enzyme Inhibitors/chemical synthesis , Humans , Models, Molecular , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutant Proteins/genetics , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/genetics , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrazoles/pharmacology , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry , Triazoles/pharmacology
13.
Assay Drug Dev Technol ; 13(2): 88-93, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25710279

ABSTRACT

Factorial experimental design (FED) is a powerful approach for efficient optimization of robust in vitro assays-it enables cost and time savings while also improving the quality of assays. Although it is a well-known technique, there can be considerable barriers to overcome to fully exploit it within an industrial or academic organization. The article describes a tactical roll out of FED to a scientist group through: training which demystifies the technical components and concentrates on principles and examples; a user-friendly Excel-based tool for deconvoluting plate data; output which focuses on graphical display of data over complex statistics. The use of FED historically has generally been in conjunction with automated technology; however we have demonstrated a much broader impact of FED on the assay development process. The standardized approaches we have rolled out have helped to integrate FED as a fundamental part of assay development best practice because it can be used independently of the automation and vendor-supplied software. The techniques are applicable to different types of assay, both enzyme and cell, and can be used flexibly in manual and automated processes. This article describes the application of FED for a cellular assay. The challenges of selling FED concepts and rolling out to a wide bioscience community together with recommendations for good working practices and effective implementation are discussed. The accessible nature of these approaches means FED can be used by industrial as well as academic users.


Subject(s)
Biological Assay/methods , Data Interpretation, Statistical , Drug Discovery , Factor Analysis, Statistical , Models, Statistical , Software , Computer Simulation , Programming Languages
14.
Mol Cancer Ther ; 14(1): 48-58, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25398829

ABSTRACT

Loss of PTEN protein results in upregulation of the PI3K/AKT pathway, which appears dependent on the PI3Kß isoform. Inhibitors of PI3Kß have potential to reduce growth of tumors in which loss of PTEN drives tumor progression. We have developed a small-molecule inhibitor of PI3Kß and PI3Kδ (AZD8186) and assessed its antitumor activity across a panel of cell lines. We have then explored the antitumor effects as single agent and in combination with docetaxel in triple-negative breast (TNBC) and prostate cancer models. In vitro, AZD8186 inhibited growth of a range of cell lines. Sensitivity was associated with inhibition of the AKT pathway. Cells sensitive to AZD8186 (GI50 < 1 µmol/L) are enriched for, but not exclusively associated with, PTEN deficiency. In vivo, AZD8186 inhibits PI3K pathway biomarkers in prostate and TNBC tumors. Scheduling treatment with AZD8186 shows antitumor activity required only intermittent exposure, and that increased tumor control is achieved when AZD8186 is used in combination with docetaxel. AZD8186 is a potent inhibitor of PI3Kß with activity against PI3Kδ signaling, and has potential to reduce growth of tumors dependent on dysregulated PTEN for growth. Moreover, AZD8186 can be combined with docetaxel, a chemotherapy commonly used to treat advanced TBNC and prostate tumors. The ability to schedule AZD8186 and maintain efficacy offers opportunity to combine AZD8186 more effectively with other drugs.


Subject(s)
Aniline Compounds/administration & dosage , Antineoplastic Agents/administration & dosage , Chromones/administration & dosage , Phosphoinositide-3 Kinase Inhibitors , Prostatic Neoplasms/drug therapy , Signal Transduction/drug effects , Taxoids/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Chromones/pharmacology , Docetaxel , Drug Synergism , Female , Humans , Male , Mice , PTEN Phosphohydrolase/deficiency , Prostatic Neoplasms/metabolism , Triple Negative Breast Neoplasms/metabolism , Xenograft Model Antitumor Assays
15.
J Med Chem ; 58(2): 943-62, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25514658

ABSTRACT

Several studies have highlighted the dependency of PTEN deficient tumors to PI3Kß activity and specific inhibition of PI3Kδ has been shown activity against human B-cell cancers. We describe the discovery and optimization of a series of 8-(1-anilino)ethyl)-2-morpholino-4-oxo-4H-chromene-6-carboxamides as PI3Kß/δ inhibitors, which led to the discovery of the clinical candidate 13, also known as AZD8186. On the basis of the lower lipophilicity of the chromen-4-one core compared to the previously utilized pyrido[1,2-a]pyrimid-4-one core, this series of compounds displayed high metabolic stability and suitable physical properties for oral administration. Compound 13 showed profound pharmacodynamic modulation of p-Akt in PTEN-deficient PC3 prostate tumor bearing mice after oral administration and showed complete inhibition of tumor growth in the mouse PTEN-deficient PC3 prostate tumor xenograft model. 13 was selected as a clinical candidate for treatment of PTEN-deficient cancers and has recently entered phase I clinical trials.


Subject(s)
Aniline Compounds/chemical synthesis , Chromones/chemical synthesis , Neoplasms, Experimental/drug therapy , PTEN Phosphohydrolase/deficiency , Phosphoinositide-3 Kinase Inhibitors , Aniline Compounds/pharmacology , Animals , Chromones/pharmacology , Dogs , Drug Discovery , Humans , Male , Mice , Neoplasms, Experimental/chemistry , Structure-Activity Relationship
16.
Bioorg Med Chem Lett ; 24(16): 3928-35, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24992874

ABSTRACT

Starting from TGX-221, we designed a series of 9-(1-anilinoethyl)-2-morpholino-4-oxo-pyrido[1,2-a]pyrimidine-7-carboxamides as potent and selective PI3Kß/δ inhibitors. Structure-activity relationships and structure-property relationships around the aniline and the amide substituents are discussed. We identified compounds 17 and 18, which showed profound pharmacodynamic modulation of phosphorylated Akt in the PC3 prostate tumour xenograft, after a single oral dose. Compound 17 also gave significant inhibition of tumour growth in the PC3 prostate tumour xenograft model after chronic oral dosing.


Subject(s)
Amides/pharmacology , Antineoplastic Agents/pharmacology , Drug Discovery , Neoplasms, Experimental/drug therapy , PTEN Phosphohydrolase/deficiency , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Administration, Oral , Amides/administration & dosage , Amides/chemistry , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Mice , Molecular Structure , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/pathology , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
17.
Bioorg Med Chem Lett ; 21(16): 4698-701, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21775140

ABSTRACT

We disclose a novel series of insulin-like growth factor-1 receptor kinase inhibitors based on the 3-(pyrimidin-4-yl)-imidazo[1,2-a]pyridine scaffold. The influence on the inhibitory activity of substitution on the imidazopyridine and at the C5 position of the pyrimidine is discussed. In the course of this optimization, we discovered a potent and selective inhibitor with suitable pharmacokinetics for oral administration.


Subject(s)
Drug Discovery , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Animals , Dogs , Humans , Mice , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyridines/chemical synthesis , Pyridines/chemistry , Rats , Stereoisomerism , Structure-Activity Relationship , Tissue Distribution
18.
Bioorg Med Chem Lett ; 18(16): 4723-6, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18676144

ABSTRACT

Tie-2 is a receptor tyrosine kinase which is involved in angiogenesis and thereby growth of human tumours. The discovery and SAR of a novel class of imidazole-vinyl-pyrimidine kinase inhibitors, which inhibit Tie-2 in vitro is reported. Their synthesis was carried out by condensation of imidazole aldehydes with methyl pyrimidines. These compounds are lead-like, with low molecular weight, good physical properties and oral bioavailability.


Subject(s)
Imidazoles/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Receptor, TIE-2/antagonists & inhibitors , Administration, Oral , Biological Availability , Chemistry, Pharmaceutical/methods , Drug Design , Humans , Imidazoles/administration & dosage , Inhibitory Concentration 50 , Models, Chemical , Molecular Conformation , Neovascularization, Pathologic , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Receptor, TIE-2/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL