Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
2.
J Med Chem ; 62(24): 11004-11018, 2019 12 26.
Article in English | MEDLINE | ID: mdl-31710489

ABSTRACT

The RAS/MAPK pathway is a major driver of oncogenesis and is dysregulated in approximately 30% of human cancers, primarily by mutations in the BRAF or RAS genes. The extracellular-signal-regulated kinases (ERK1 and ERK2) serve as central nodes within this pathway. The feasibility of targeting the RAS/MAPK pathway has been demonstrated by the clinical responses observed through the use of BRAF and MEK inhibitors in BRAF V600E/K metastatic melanoma; however, resistance frequently develops. Importantly, ERK1/2 inhibition may have clinical utility in overcoming acquired resistance to RAF and MEK inhibitors, where RAS/MAPK pathway reactivation has occurred, such as relapsed BRAF V600E/K melanoma. We describe our structure-based design approach leading to the discovery of AZD0364, a potent and selective inhibitor of ERK1 and ERK2. AZD0364 exhibits high cellular potency (IC50 = 6 nM) as well as excellent physicochemical and absorption, distribution, metabolism, and excretion (ADME) properties and has demonstrated encouraging antitumor activity in preclinical models.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Discovery , Imidazoles/therapeutic use , Lung Neoplasms/drug therapy , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrazines/therapeutic use , Pyrimidines/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Apoptosis , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation , Drug Therapy, Combination , Female , Humans , Imidazoles/pharmacology , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice , Mice, Nude , Molecular Structure , Protein Kinase Inhibitors/administration & dosage , Pyrazines/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/therapeutic use , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
J Med Chem ; 62(14): 6540-6560, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31199640

ABSTRACT

Tumors have evolved a variety of methods to reprogram conventional metabolic pathways to favor their own nutritional needs, including glutaminolysis, the first step of which is the hydrolysis of glutamine to glutamate by the amidohydrolase glutaminase 1 (GLS1). A GLS1 inhibitor could potentially target certain cancers by blocking the tumor cell's ability to produce glutamine-derived nutrients. Starting from the known GLS1 inhibitor bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide, we describe the medicinal chemistry evolution of a series from lipophilic inhibitors with suboptimal physicochemical and pharmacokinetic properties to cell potent examples with reduced molecular weight and lipophilicity, leading to compounds with greatly improved oral exposure that demonstrate in vivo target engagement accompanied by activity in relevant disease models.


Subject(s)
Antineoplastic Agents/pharmacology , Glutaminase/antagonists & inhibitors , Neoplasms/drug therapy , Pyridazines/pharmacology , Thiadiazoles/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Biological Availability , Cell Line, Tumor , Drug Discovery , Glutaminase/metabolism , Humans , Male , Mice, SCID , Molecular Docking Simulation , Neoplasms/metabolism , Neoplasms/pathology , Pyridazines/chemistry , Pyridazines/pharmacokinetics , Pyridazines/therapeutic use , Thiadiazoles/chemistry , Thiadiazoles/pharmacokinetics , Thiadiazoles/therapeutic use
4.
Nat Rev Drug Discov ; 17(3): 167-181, 2018 03.
Article in English | MEDLINE | ID: mdl-29348681

ABSTRACT

In 2011, AstraZeneca embarked on a major revision of its research and development (R&D) strategy with the aim of improving R&D productivity, which was below industry averages in 2005-2010. A cornerstone of the revised strategy was to focus decision-making on five technical determinants (the right target, right tissue, right safety, right patient and right commercial potential). In this article, we describe the progress made using this '5R framework' in the hope that our experience could be useful to other companies tackling R&D productivity issues. We focus on the evolution of our approach to target validation, hit and lead optimization, pharmacokinetic/pharmacodynamic modelling and drug safety testing, which have helped improve the quality of candidate drug nomination, as well as the development of the right culture, where 'truth seeking' is encouraged by more rigorous and quantitative decision-making. We also discuss where the approach has failed and the lessons learned. Overall, the continued evolution and application of the 5R framework are beginning to have an impact, with success rates from candidate drug nomination to phase III completion improving from 4% in 2005-2010 to 19% in 2012-2016.


Subject(s)
Biomedical Research/standards , Decision Making, Organizational , Drug Industry , Drugs, Investigational/therapeutic use , Efficiency, Organizational , Research Design , Research/organization & administration , Clinical Trials as Topic , Efficiency , Humans , Organizational Culture , Research/standards
5.
J Med Chem ; 57(20): 8249-67, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25271963

ABSTRACT

Epidermal growth factor receptor (EGFR) inhibitors have been used clinically in the treatment of non-small-cell lung cancer (NSCLC) patients harboring sensitizing (or activating) mutations for a number of years. Despite encouraging clinical efficacy with these agents, in many patients resistance develops leading to disease progression. In most cases, this resistance is in the form of the T790M mutation. In addition, EGFR wild type receptor inhibition inherent with these agents can lead to dose limiting toxicities of rash and diarrhea. We describe herein the evolution of an early, mutant selective lead to the clinical candidate AZD9291, an irreversible inhibitor of both EGFR sensitizing (EGFRm+) and T790M resistance mutations with selectivity over the wild type form of the receptor. Following observations of significant tumor inhibition in preclinical models, the clinical candidate was administered clinically to patients with T790M positive EGFR-TKI resistant NSCLC and early efficacy has been observed, accompanied by an encouraging safety profile.


Subject(s)
Acrylamides/pharmacology , Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/genetics , Chemistry Techniques, Synthetic , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/genetics , Female , Humans , Inhibitory Concentration 50 , Lung Neoplasms/genetics , Male , Mice , Middle Aged , Mutation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Rats, Inbred Strains , Xenograft Model Antitumor Assays
6.
Genomics ; 104(2): 96-104, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25043885

ABSTRACT

Long interspersed nuclear elements (Line-1 or L1s) account for ~17% of the human genome. While the majority of human L1s are inactive, ~80-100 elements remain retrotransposition competent and mobilize through RNA intermediates to different locations within the genome. De novo insertions of L1s account for polymorphic variation of the human genome and disruption of target loci at their new location. In the present study, fluorescence in situ hybridization and DNA sequencing were used to characterize retrotransposition profiles of L1(RP) in cultured human HepG2 cells. While expression of synthetic L1(RP) was associated with full-length and truncated insertions throughout the entire genome, a strong preference for gene-poor regions, such as those found in chromosome 13 was observed for full-length insertions. These findings shed light into L1 targeting mechanisms within the human genome and question the putative randomness of L1 retrotransposition.


Subject(s)
Chromosomes, Human, Pair 13/genetics , Genome, Human , Long Interspersed Nucleotide Elements/genetics , Base Sequence , Cloning, Molecular , Hep G2 Cells , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Mutagenesis, Insertional , Plasmids/genetics , Sequence Analysis, DNA
7.
J Med Chem ; 56(17): 7025-48, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-23930994

ABSTRACT

A novel series of small-molecule inhibitors has been developed to target the double mutant form of the epidermal growth factor receptor (EGFR) tyrosine kinase, which is resistant to treatment with gefitinib and erlotinib. Our reported compounds also show selectivity over wild-type EGFR. Guided by molecular modeling, this series was evolved to target a cysteine residue in the ATP binding site via covalent bond formation and demonstrates high levels of activity in cellular models of the double mutant form of EGFR. In addition, these compounds show significant activity against the activating mutations, which gefitinib and erlotinib target and inhibition of which gives rise to their observed clinical efficacy. A glutathione (GSH)-based assay was used to measure thiol reactivity toward the electrophilic functionality of the inhibitor series, enabling both the identification of a suitable reactivity window for their potency and the development of a reactivity quantitative structure-property relationship (QSPR) to support design.


Subject(s)
ErbB Receptors/antagonists & inhibitors , ErbB Receptors/chemistry , ErbB Receptors/genetics , Models, Molecular , Mutation , Structure-Activity Relationship
8.
Mol Oncol ; 7(4): 812-25, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23648019

ABSTRACT

Long Interspersed Nuclear Element-1 (LINE-1 or L1) is an autonomous, mobile element within the human genome that transposes via a "copy and paste" mechanism and relies upon L1-encoded endonuclease and reverse transcriptase (RT) activities to compromise genome integrity. L1 has been implicated in various forms of cancer, but its role in the regulation of the oncogenic phenotype is not understood. The present studies were conducted to evaluate mechanisms of genetic regulatory control in HepG2 cells by human L1, or a D702Y mutant deficient in RT activity, and their influence on cellular phenotype. Forced expression of synthetic L1 ORF1p and ORF2p was associated with formation of cytoplasmic foci and minor association with the nuclear compartment. While de novo L1 mobilizations were only identified in cells expressing wild type L1, and were absent in the D702Y mutant, changes in gene expression profiles involved RT dependent as well as RT independent mechanisms. Synthetic L1 altered the expression of 24 in silico predicted genetic targets; ten of which showed RT-dependence, ten RT-independence, and four reciprocal regulatory control by both wild type and RT mutant. Of five targets examined, only VCAM1 and PTPRB colocalized with newly retrotransposed wild type L1. Biological discretization to partition patterns of gene expression into unique frequencies identified adhesion, inflammation, and cellular metabolism as key processes targeted for molecular interference with disruption of epithelial-to-mesenchymal programming seen irrespective of the RT phenotype. These findings establish L1 as a key regulator of genome plasticity and EMT via mechanisms independent of RT activity.


Subject(s)
Long Interspersed Nucleotide Elements/genetics , Retroelements/genetics , Fluorescent Antibody Technique, Indirect , Hep G2 Cells , Humans , In Situ Hybridization, Fluorescence
9.
Toxicol Pathol ; 39(6): 916-24, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21859884

ABSTRACT

Aberrant signaling by transforming growth factor-ß (TGF-ß) and its type I (ALK5) receptor has been implicated in a number of human diseases and this pathway is considered a potential target for therapeutic intervention. Transforming growth factor-ß signaling via ALK5 plays a critical role during heart development, but the role of ALK5 in the adult heart is poorly understood. In the current study, the preclinical toxicology of ALK5 inhibitors from two different chemistry scaffolds was explored. Ten-week-old female Han Wistar rats received test compounds by the oral route for three to seven days. Both compounds induced histopathologic heart valve lesions characterized by hemorrhage, inflammation, degeneration, and proliferation of valvular interstitial cells. The pathology was observed in all animals, at all doses tested, and occurred in all four heart valves. Immunohistochemical analysis of ALK5 in rat hearts revealed expression in the valves, but not in the myocardium. Compared to control animals, protein levels of ALK5 were unchanged in the heart valves of treated animals. We also observed a physeal dysplasia in the femoro-tibial joint of rats treated with ALK5 inhibitors, a finding consistent with a pharmacological effect described previously with ALK5 inhibitors. Overall, these findings suggest that TGF-ß signaling via ALK5 plays a critical role in maintaining heart valve integrity.


Subject(s)
Heart Valves/pathology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , Administration, Oral , Animals , Drug Evaluation, Preclinical , Female , Heart Valves/drug effects , Immunohistochemistry/methods , Protein Serine-Threonine Kinases/genetics , Rats , Rats, Wistar , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Transforming Growth Factor beta/genetics
10.
Clin Cancer Res ; 10(15): 5233-41, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15297427

ABSTRACT

Indole-3-carbinol (I3C) and 3,3'-diindolylmethane (DIM) are promising cancer chemopreventive agents in rodent models, but there is a paucity of data on their pharmacokinetics and tissue disposition. The disposition of I3C and its acid condensation products, DIM, [2-(indol-3-ylmethyl)-indol-3-yl]indol-3-ylmethane (LTr(1)), indolo[3,2b]carbazole (ICZ) and 1-(3-hydroxymethyl)-indolyl-3-indolylmethane (HI-IM) was studied, after oral administration of I3C (250 mg/kg) to female CD-1 mice. Blood, liver, kidney, lung, heart, and brain were collected between 0.25 and 24 h after administration and the plasma and tissue concentrations of I3C and its derivatives determined by high-performance liquid chromotography. I3C was rapidly absorbed, distributed, and eliminated from plasma and tissues, falling below the limit of detection by 1 h. Highest concentrations of I3C were detected in the liver where levels were approximately 6-fold higher than those in the plasma. Levels of DIM, LTr(1), and HI-IM were much lower, although they persisted in plasma and tissues for considerably longer. DIM and HI-IM were still present in the liver 24 h after I3C administration. Tissue levels of DIM and LTr(1) were found to be in equilibrium with plasma at almost every time point measured. In addition to acid condensation products of I3C, a major oxidative metabolite (indole-3-carboxylic acid) and a minor oxidative metabolite (indole-3-carboxaldehyde) were detected in plasma of mice after oral administration of I3C. ICZ was also tentatively identified in the liver of these mice. This study shows for the first time that, after oral administration to mice, I3C, in addition to its acid condensation products, is absorbed from the gut and distributed systemically into a number of well-perfused tissues, thus allowing the possibility for some pharmacological activity of the parent compound in vivo.


Subject(s)
Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/pharmacokinetics , Indoles/administration & dosage , Indoles/pharmacokinetics , Administration, Oral , Animals , Anticarcinogenic Agents/blood , Chromatography, High Pressure Liquid , Chromatography, Liquid , Female , Free Radical Scavengers/administration & dosage , Free Radical Scavengers/blood , Free Radical Scavengers/pharmacokinetics , Indoles/blood , Liver/metabolism , Mice , Models, Chemical , Oxygen/metabolism , Perfusion , Time Factors , Tissue Distribution
11.
Drug Metab Dispos ; 32(6): 632-8, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15155555

ABSTRACT

3,3'-Diindolylmethane (DIM) is a naturally occurring indole, which is currently under investigation as a potential chemopreventive agent. The concentrations of DIM in plasma, liver, kidney, lung, heart, and brain tissues were determined following oral administration of two different formulations to mice (250 mg/kg). Mice were sacrificed periodically from 0 to 24 h after administration of either a crystalline or an absorption-enhanced formulation (Bio-Response-DIM; Indolplex) of DIM, and plasma and tissue concentrations were determined by high-performance liquid chromatography (UV detection, 280 nm). A physiologically based pharmacokinetic (PBPK) model was developed to characterize the pharmacokinetic properties of the two different formulations. The final model included parameters reflecting linear first-order absorption, systemic clearance, and distributional clearance in the remainder compartment, which were considered independent of formulation. All pharmacokinetic profiles from the two formulations were fitted simultaneously to estimate unknown model parameters. Plasma and tissue concentration-time profiles exhibited a rapid rise to peak values at 0.5 to 1 h, followed by a polyexponential decline with an extended terminal phase. These profiles were well described by the final model and unknown parameters were estimated with relatively low coefficients of variation. Relative drug exposure and absorption parameters suggest that BioResponse-DIM exhibited approximately 50% higher bioavailability than the crystalline formulation. Clearance of DIM was estimated as 7.18 ml/h. This is the first study to characterize the pharmacokinetics of DIM in mice, and the established PBPK model should prove useful in the design and analysis of future preclinical studies aimed at evaluating the in vivo pharmacological effects of DIM.


Subject(s)
Anticarcinogenic Agents/pharmacokinetics , Indoles/pharmacokinetics , Administration, Oral , Animals , Anticarcinogenic Agents/blood , Area Under Curve , Chemistry, Pharmaceutical , Chromatography, Liquid , Female , Indoles/blood , Mice , Mice, Inbred Strains , Models, Biological , Tissue Distribution
12.
Article in English | MEDLINE | ID: mdl-12650751

ABSTRACT

A high-performance liquid chromatographic method was developed for the simultaneous determination of indole-3-carbinol (I3C), 3,3'-diindolylmethane (DIM), [2-(indol-3-ylmethyl)-indol-3-yl]indol-3-ylmethane (LTr(1)), and indolo[3,2b]carbazole (ICZ). Compounds were extracted from mouse plasma using tert.-butyl methyl ether, incorporating 4-methoxy-indole as internal standard. Chromatographic separation utilized a Waters Symmetry RP18 in tandem with a Thermoquest BDS C(18) column, an acetonitrile-water gradient and UV (280 nm) in series with fluorescence (ex. 335 nm; em. 415 nm) detection. Calibration curves were linear (r(2)>0.99) between 50 and 15,000 ng/ml for I3C; 150 and 15,000 ng/ml for LTr(1); and 0.15 and 37.5 ng/ml for ICZ and the method was reproducible and precise (within-day and between-day coefficients of variation below 9.7 and 13%, respectively). The method described is suitable for comprehensive pharmacokinetic studies with indole-3-carbinol.


Subject(s)
Chromatography, High Pressure Liquid/methods , Indoles/blood , Calibration , Reference Standards , Reproducibility of Results , Sensitivity and Specificity , Spectrophotometry, Ultraviolet
13.
Brain Res Mol Brain Res ; 102(1-2): 62-72, 2002 Jun 15.
Article in English | MEDLINE | ID: mdl-12191495

ABSTRACT

The signalling pathways by which muscarine and epidermal growth factor (EGF) regulate the secretion of the alpha-secretase cleavage product (sAPPalpha) of the amyloid precursor protein (APP) were examined in the human neuroblastoma SH-SY5Y. Using specific inhibitors it was found that over 80% of sAPPalpha secretion, enhanced by muscarine, occurred via the extracellular signal-regulated kinase (ERK1/2) member of the mitogen-activated protein kinase (MAPK) family and was dependent on protein kinase Calpha (PKCalpha) and a member of the Src family of non-receptor tyrosine kinases (Src-TK). In contrast the stimulation of sAPPalpha secretion by EGF was not affected by inhibitors of PKC nor Src-TK but was dependent on ERK1/2. In addition muscarine-enhanced sAPPalpha secretion and ERK1/2 activation were inhibited 60 and 80%, respectively, by micromolar concentrations of the phosphatidylinositol 3 kinase (PI-3K) inhibitor wortmannin. In comparison wortmannin decreased EGF stimulation of sAPPalpha secretion and ERK 1/2 activation by approximately 40%. Unexpectedly, U73122, an inhibitor of phosphoinositide-specific phospholipase C, did not inhibit muscarine enhancement of sAPPalpha secretion. These data are discussed in relation to a pathway for the enhancement of sAPPalpha secretion by muscarine which involves the activation of a Src-TK by G-protein beta/gamma-subunits leading to activation of PKCalpha, and ERK1/2 by a mechanism not involving phospholipase C.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Protein Precursor/biosynthesis , Gene Expression Regulation, Enzymologic/physiology , Muscarine/pharmacology , Muscarinic Agonists/pharmacology , Neurons/enzymology , Signal Transduction/physiology , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/drug effects , Amyloid beta-Protein Precursor/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Humans , Isoenzymes/drug effects , Isoenzymes/metabolism , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Neuroblastoma , Neurons/drug effects , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Protein Kinase C-alpha , Signal Transduction/drug effects , Tumor Cells, Cultured , Type C Phospholipases/drug effects , Type C Phospholipases/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology , src-Family Kinases/drug effects , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...