Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Med Chem ; 65(5): 4350-4366, 2022 03 10.
Article in English | MEDLINE | ID: mdl-35195996

ABSTRACT

Mutations in MEK1/2 have been described as a resistance mechanism to BRAF/MEK inhibitor treatment. We report the discovery of a novel ATP-competitive MEK1/2 inhibitor with efficacy in wildtype (WT) and mutant MEK12 models. Starting from a HTS hit, we obtained selective, cellularly active compounds that showed equipotent inhibition of WT MEK1/2 and a panel of MEK1/2 mutant cell lines. Using a structure-based approach, the optimization addressed the liabilities by systematic analysis of molecular matched pairs (MMPs) and ligand conformation. Addition of only three heavy atoms to early tool compound 6 removed Cyp3A4 liabilities and increased the cellular potency by 100-fold, while reducing log P by 5 units. Profiling of MAP855, compound 30, in pharmacokinetic-pharmacodynamic and efficacy studies in BRAF-mutant models showed comparable efficacy to clinical MEK1/2 inhibitors. Compound 30 is a novel highly potent and selective MEK1/2 kinase inhibitor with equipotent inhibition of WT and mutant MEK1/2, whose drug-like properties allow further investigation in the mutant MEK setting upon BRAF/MEK therapy.


Subject(s)
Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Adenosine Triphosphate/metabolism , Cell Line, Tumor , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics
2.
ChemMedChem ; 14(13): 1238-1247, 2019 07 03.
Article in English | MEDLINE | ID: mdl-30957954

ABSTRACT

Histamine H3 receptor (H3R) inverse agonists that have been in clinical trials for the treatment of excessive sleep disorders, have been plagued with insomnia as a mechanism-based side effect. We focused on the identification of compounds that achieve high receptor occupancy within a short time, followed by rapid disengagement from the receptor, a target profile that could provide therapeutic benefits without the undesired side effect of insomnia. This article describes the optimization work that led to the discovery of 1-(1-methyl-6-oxo-1,6-dihydropyridazin-3-yl)piperidin-4-yl 4-cyclobutylpiperazine-1-carboxylate (18 b, LML134).


Subject(s)
Histamine Agonists/therapeutic use , Piperazine/chemistry , Piperazines/chemistry , Receptors, Histamine H3/metabolism , Sleep Wake Disorders/drug therapy , Animals , Drug Evaluation, Preclinical , Drug Inverse Agonism , Half-Life , Histamine Agonists/chemistry , Histamine Agonists/pharmacokinetics , Humans , Male , Microsomes, Liver/metabolism , Piperazine/pharmacokinetics , Piperazine/therapeutic use , Piperazines/pharmacokinetics , Piperazines/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Histamine H3/chemistry , Structure-Activity Relationship
3.
ChemMedChem ; 10(2): 266-75, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25394333

ABSTRACT

Ergolines were recently identified as a novel class of H3 receptor (H3R) inverse agonists. Although their optimization led to drug candidates with encouraging properties for the treatment of narcolepsy, brain penetration remained low. To overcome this issue, ergoline 1 ((6aR,9R,10aR)-4-(2-(dimethylamino)ethyl)-N-phenyl-9-(pyrrolidine-1-carbonyl)-6,6a,8,9,10,10a-hexahydroindolo[4,3-fg]quinoline-7(4H)-carboxamide)) was transformed into a series of indole derivatives with high H3R affinity. These new molecules were profiled by simultaneous determination of their brain receptor occupancy (RO) levels and pharmacodynamic (PD) effects in mice. These efforts culminated in the discovery of 15 m ((R)-1-isopropyl-5-(1-(2-(2-methylpyrrolidin-1-yl)ethyl)-1H-indol-4-yl)pyridin-2(1H)-one), which has an ideal profile showing a strong correlation of PD effects with RO, and no measurable safety liabilities. Its desirably short duration of action was confirmed by electroencephalography (EEG) measurements in rats.


Subject(s)
Ergolines/chemistry , Histamine Antagonists/chemistry , Indoles/chemistry , Pyridones/chemistry , Receptors, Histamine H3/chemistry , Animals , Brain/metabolism , CHO Cells , Cricetinae , Cricetulus , Electroencephalography , Ergolines/pharmacokinetics , Ergolines/therapeutic use , Half-Life , Histamine Antagonists/pharmacokinetics , Histamine Antagonists/therapeutic use , Humans , Indoles/pharmacokinetics , Indoles/therapeutic use , Male , Mice , Narcolepsy/drug therapy , Narcolepsy/metabolism , Narcolepsy/pathology , Protein Binding , Pyridones/pharmacokinetics , Pyridones/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Histamine H3/metabolism , Structure-Activity Relationship
4.
J Med Chem ; 57(17): 7396-411, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25121964

ABSTRACT

We describe the synthesis and characterization of 3-alkoxy-pyrrolo[1,2-b]pyrazolines as novel selective androgen receptor (AR) modulators that possess excellent physicochemical properties for transdermal administration. Compound 26 bound to human AR with an IC50 of 0.7 nM with great selectivity over other nuclear hormone receptors and potently activated AR in a C2C12 muscle cell reporter gene assay with an EC50 of 0.5 nM. It showed high aqueous solubility of 1.3 g/L at pH 7.4, and an in silico model as well as a customized parallel artificial membrane permeability assay indicated good skin permeation. Indeed, when measuring skin permeation through excised human skin, an excellent flux of 2 µg/(cm(2)·h) was determined without any permeation enhancers. In a 2 week Hershberger model using castrated rats, the compound showed dose-dependent effects fully restoring skeletal muscle weight at 0.3 mg/kg/day after subcutaneous administration with high selectivity over prostate stimulation.


Subject(s)
Androgen Receptor Antagonists/chemistry , Androgens/chemistry , Azabicyclo Compounds/chemistry , Pyrazoles/chemistry , Receptors, Androgen/chemistry , Administration, Cutaneous , Androgen Receptor Antagonists/metabolism , Androgen Receptor Antagonists/pharmacokinetics , Androgens/metabolism , Animals , Area Under Curve , Azabicyclo Compounds/metabolism , Azabicyclo Compounds/pharmacokinetics , Binding Sites , Binding, Competitive , Cell Line , Chemical Phenomena , Crystallography, X-Ray , Humans , Male , Metabolic Clearance Rate , Mice , Models, Chemical , Models, Molecular , Molecular Structure , Protein Structure, Tertiary , Pyrazoles/metabolism , Pyrazoles/pharmacokinetics , Rats, Wistar , Receptors, Androgen/metabolism , Skin/metabolism
5.
ChemMedChem ; 9(8): 1683-96, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24850792

ABSTRACT

Ergoline derivative (6aR,9R)-4-(2-(dimethylamino)ethyl)-N-phenyl-9-(pyrrolidine-1-carbonyl)-6,6a,8,9-tetrahydroindolo[4,3-fg]quinoline-7(4H)-carboxamide (1), a CXCR3 antagonist, also inhibits human histamine H3 receptors (H3R) and represents a structurally novel H3R inverse agonist chemotype. It displays favorable pharmacokinetic and in vitro safety profiles, and served as a lead compound in a program to explore ergoline derivatives as potential drug candidates for the treatment of narcolepsy. A key objective of this work was to enhance the safety and efficacy profiles of 1, while minimizing its duration of action to mitigate the episodes of insomnia documented with previously reported clinical candidates during the night following administration. Modifications to the ergoline core at positions 1, 6 and 8 were systematically investigated, and derivative 23 (1-((4aR,8R,9aR)-8-(hydroxymethyl)-1-(2-((R)-2-methylpyrrolidin-1-yl)ethyl)-4,4a,7,8,9,9a-hexahydroindolo[1,14-fg]quinolin-6(1H)-yl)ethanone) was identified as a promising lead compound. Derivative 23 has a desirable pharmacokinetic profile and demonstrated efficacy by enhancing brain concentrations of tele-methylhistamine, a major histamine metabolite. This validates the potential of the ergoline scaffold to serve as a template for the development of H3R inverse agonists.


Subject(s)
Ergolines/chemistry , Histamine Agonists/chemistry , Receptors, Histamine H3/chemistry , Animals , Caco-2 Cells , Cell Line , Dogs , Drug Inverse Agonism , Ergolines/pharmacokinetics , Ergolines/therapeutic use , Half-Life , Histamine Agonists/pharmacokinetics , Histamine Agonists/therapeutic use , Humans , Madin Darby Canine Kidney Cells , Male , Mice , Microsomes, Liver/metabolism , Narcolepsy/drug therapy , Protein Binding , Rats , Rats, Sprague-Dawley , Receptors, Histamine H3/metabolism , Structure-Activity Relationship
6.
Expert Opin Ther Targets ; 8(6): 587-96, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15584864

ABSTRACT

The cataloguing of the human genome has provided an unprecedented prospectus for target identification and drug discovery. A current analysis indicates that slightly more than 3000 unique protein encoding loci are potentially amenable to pharmacological intervention (the 'druggable genome', which can be queried at http://function.gnf.org/druggable). However, the assessment of genome sequence data has not resulted in the anticipated acceleration of novel therapeutic developments. The basis for this shortfall lies in the significant attrition rates endemic to preclinical/clinical development, as well as the often underestimated complexity of gene function in higher order biological systems. To address the latter issue, a number of strategies have emerged to facilitate genomics-driven target identification and validation, including cellular profiling of gene function, in silico modelling of gene networks, and systematic analyses of protein complexes. The expectation is that the integration of these and other systems-based technologies may enable the conversion of potential genomic targets into functionally validated molecules, and result in practicable gene-based drug discovery pipelines.


Subject(s)
Drug Design , Genomics , Animals , Drug Evaluation, Preclinical , Forecasting , Gene Expression Profiling , Gene Targeting , Genome, Human , Humans , Ion Channels/drug effects , Mice , Mice, Knockout , Models, Animal , Multigene Family/drug effects , Protease Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Structure-Activity Relationship
7.
J Pharmacol Exp Ther ; 309(1): 275-84, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14718594

ABSTRACT

Angiotensin II and endothelin-1 activate their respective AT(1) and ET(A) receptors on vascular smooth muscle cells, producing vasoconstriction, and both peptides are implicated in the pathogenesis of essential hypertension. Angiotensin II potentiates the production of endothelin, and conversely endothelin augments the synthesis of angiotensin II. Both AT(1) and ET(A) receptor antagonists lower blood pressure in hypertensive patients; thus, a combination AT(1)/ET(A) receptor antagonist may have greater efficacy and broader utility compared with each drug alone. By rational drug design a biphenyl ET(A) receptor blocker was modified to acquire AT(1) receptor antagonism. These compounds (C and D) decreased Sar-Ile-Angiotensin II binding to AT(1) receptors and endothelin-1 binding to ET(A) receptors, and compound C inhibited angiotensin II- and endothelin-1-mediated Ca(2+) transients. In rats compounds C and D reduced blood pressure elevations caused by intravenous infusion of angiotensin II or big endothelin-1. Compound C decreased blood pressure in Na(+)-depleted spontaneously hypertensive rats and in rats with mineralocorticoid hypertension. Compound D was more efficacious than AT(1) receptor antagonists at reducing blood pressure in spontaneously hypertensive rats, and its superiority was likely due to its partial blockade of ET(A) receptors. Therefore compounds C and D are novel agents for treating a broad spectrum of patients with essential hypertension and other cardiovascular diseases.


Subject(s)
Antihypertensive Agents/therapeutic use , Biphenyl Compounds/therapeutic use , Blood Pressure/drug effects , Hypertension/drug therapy , Oxazoles/therapeutic use , Receptor, Angiotensin, Type 1/metabolism , Receptor, Endothelin A/metabolism , Sulfonamides/therapeutic use , Angiotensin II Type 1 Receptor Blockers , Animals , Antihypertensive Agents/pharmacology , Biphenyl Compounds/pharmacology , Calcium/metabolism , Desoxycorticosterone , Disease Models, Animal , Endothelin A Receptor Antagonists , Humans , Irbesartan , Losartan/therapeutic use , Male , Oxazoles/pharmacology , Rats , Rats, Inbred SHR , Sodium/metabolism , Sulfonamides/pharmacology , Tetrazoles/pharmacology , Tetrazoles/therapeutic use
9.
J Cardiovasc Pharmacol ; 41(6): 817-29, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12775958

ABSTRACT

Activities of vascular smooth muscle cells (SMCs) such as proliferation, migration, and matrix production contribute to restenosis following clinical interventions of angioplasty and stent placement. Because activation of platelet-derived growth factor (PDGF)-receptor tyrosine kinase (PDGFr-TK) influences these processes and promotes restenosis, TKI963, an inhibitor of the PDGFr-TK was discovered, and its efficacy was evaluated in blocking stent-induced restenosis as analyzed by intravascular ultrasound (IVUS). TKI963, a low-molecular-weight compound, inhibited the cell-free PDGFbetar-TK with a K(i) value of 56 +/- 14 nM. TKI963 also inhibited PDGF-dependent events in human aortic SMCs (e.g., in situ PDGFr autophosphorylation, mitogenesis, chemotaxis, and collagen production with median inhibitory concentration values of approximately 300 nM) without affecting the activity of a series of membrane receptor tyrosine kinases and intracellular serine/threonine kinases. In vivo, stent-induced restenosis in the swine coronary artery was reduced by oral administration of TKI963 (1.25, 2.5, and 5 mg/kg BID, for 28 days). Late lumen cross-sectional area (CSA) loss, plaque CSA growth, and plaque volume in the stent determined by IVUS were dose-relatedly decreased (33-62% at 1.25 mg/kg BID to 66-92% at 5 mg/kg BID, depending on the parameter) compared with controls. TKI963 treatment of

Subject(s)
Coronary Restenosis/etiology , Coronary Restenosis/prevention & control , Coronary Vessels/drug effects , Quinoxalines/pharmacology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Stents/adverse effects , Administration, Oral , Animals , Cell Division/drug effects , Cells, Cultured , Chemotaxis/drug effects , Collagen/biosynthesis , Coronary Vessels/metabolism , Coronary Vessels/pathology , Male , Mice , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Phosphorylation , Rats , Receptors, Platelet-Derived Growth Factor/metabolism , Swine, Miniature
SELECTION OF CITATIONS
SEARCH DETAIL