Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Eur J Med Chem ; 260: 115725, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37657269

ABSTRACT

This paper describes the rational design, synthesis, structure-activity relationship (SAR), and biological profile of presenilin-1 (PSEN-1) complex selective γ-secretase inhibitors, assessed for selectivity using a unique set of four γ-secretase subtype complexes. A set of known PSEN-1 selective γ-Secretase inhibitors (GSIs) was analyzed to understand the pharmacophoric features required for selective inhibition. Conformational modeling suggests that a characteristic 'U' shape orientation between aromatic sulfone/sulfonamide and aryl ring is crucial for PSEN-1 selectivity and potency. Using these insights, a series of brain-penetrant 2-azabicyclo[2,2,2]octane sulfonamides was devised and synthesized as a new class of PSEN-1 selective inhibitors. Compounds 13c and 13k displayed high potency towards PSEN1-APH1B complex but moderate selectivity towards PSEN2 complexes. However, compound (+)-13b displayed low nanomolar potency towards the PSEN1-APH1B complex, little (∼4-fold) selectivity towards PSEN1-APH1A, and high selectivity (>350-fold) versus PSEN2 complexes. Excellent brain penetration, no significant CYP inhibition, or cardiotoxicity, good solubility, and permeability make (+)-13b an excellent candidate for further lead optimization.


Subject(s)
Amyloid Precursor Protein Secretases , Sulfonamides , Sulfonamides/pharmacology , Presenilin-1 , Octanes , Sulfanilamide , Brain
2.
J Biol Chem ; 299(6): 104794, 2023 06.
Article in English | MEDLINE | ID: mdl-37164155

ABSTRACT

Clinical development of γ-secretases, a family of intramembrane cleaving proteases, as therapeutic targets for a variety of disorders including cancer and Alzheimer's disease was aborted because of serious mechanism-based side effects in the phase III trials of unselective inhibitors. Selective inhibition of specific γ-secretase complexes, containing either PSEN1 or PSEN2 as the catalytic subunit and APH1A or APH1B as supporting subunits, does provide a feasible therapeutic window in preclinical models of these disorders. We explore here the pharmacophoric features required for PSEN1 versus PSEN2 selective inhibition. We synthesized a series of brain penetrant 2-azabicyclo[2,2,2]octane sulfonamides and identified a compound with low nanomolar potency and high selectivity (>250-fold) toward the PSEN1-APH1B subcomplex versus PSEN2 subcomplexes. We used modeling and site-directed mutagenesis to identify critical amino acids along the entry part of this inhibitor into the catalytic site of PSEN1. Specific targeting one of the different γ-secretase complexes might provide safer drugs in the future.


Subject(s)
Amyloid Precursor Protein Secretases , Multiprotein Complexes , Presenilin-1 , Sulfonamides , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Presenilin-1/antagonists & inhibitors , Presenilin-1/metabolism , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/metabolism , Sulfonamides/pharmacology , Substrate Specificity , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/metabolism
3.
ChemMedChem ; 14(22): 1894-1910, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31657130

ABSTRACT

The ß-site amyloid precursor protein cleaving enzyme 1 (BACE1, also known as ß-secretase) is a promising target for the treatment of Alzheimer's disease. A pKa lowering approach over the initial leads was adopted to mitigate hERG inhibition and P-gp efflux, leading to the design of 6-CF3 dihydrothiazine 8 (N-(3-((4S,6S)-2-amino-4-methyl-6-(trifluoromethyl)-5,6-dihydro-4H-1,3-thiazin-4-yl)-4-fluorophenyl)-5-cyanopicolinamide). Optimization of 8 led to the discovery of 15 (N-(3-((4S,6S)-2-amino-4-methyl-6-(trifluoromethyl)-5,6-dihydro-4H-1,3-thiazin-4-yl)-4-fluorophenyl)-5-(fluoromethoxy)pyrazine-2-carboxamide) with an excellent balance of potency, hERG inhibition, P-gp efflux, and metabolic stability. Oral administration of 8 elicited robust Aß reduction in dog even at 0.16 mg/kg. Reflecting the reduced hERG inhibitory activity, no QTc prolongation was observed at high doses. The potential for reactive metabolite formation of 15 was realized in a nucleophile trapping assay using [14 C]-KCN in human liver microsomes. Utilizing covalent binding (CVB) in human hepatocytes and the maximum projected human dosage, the daily CVB burden of 15 was calculated to be at an acceptable value of below 1 mg/day. However, hepatotoxicity was observed when 15 was subjected to a two-week tolerance study in dog, which prevented further evaluation of this compound.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Oxazines/pharmacology , Thiazines/pharmacology , Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/deficiency , Aspartic Acid Endopeptidases/metabolism , Dogs , Dose-Response Relationship, Drug , Drug Design , Hepatocytes/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Models, Molecular , Molecular Structure , Oxazines/chemistry , Rats , Structure-Activity Relationship , Thiazines/administration & dosage , Thiazines/chemistry
4.
EMBO J ; 38(12)2019 06 17.
Article in English | MEDLINE | ID: mdl-31109937

ABSTRACT

γ-Secretase complexes (GSECs) are multimeric membrane proteases involved in a variety of physiological processes and linked to Alzheimer's disease (AD). Presenilin (PSEN, catalytic subunit), Nicastrin (NCT), Presenilin Enhancer 2 (PEN-2), and Anterior Pharynx Defective 1 (APH1) are the essential subunits of GSECs. Mutations in PSEN and the Amyloid Precursor Protein (APP) cause early-onset AD GSECs successively cut APP to generate amyloid-ß (Aß) peptides of various lengths. AD-causing mutations destabilize GSEC-APP/Aßn interactions and thus enhance the production of longer Aßs, which elicit neurotoxic events underlying pathogenesis. Here, we investigated the molecular strategies that anchor GSEC and APP/Aßn during the sequential proteolysis. Our studies reveal that a direct interaction between NCT ectodomain and APPC99 influences the stability of GSEC-Aßn assemblies and thereby modulates Aß length. The data suggest a potential link between single-nucleotide variants in NCSTN and AD risk. Furthermore, our work indicates that an extracellular interface between the protease (NCT, PSEN) and the substrate (APP) represents the target for compounds (GSMs) modulating Aß length. Our findings may guide future rationale-based drug discovery efforts.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Membrane Glycoproteins/metabolism , Protein Interaction Domains and Motifs/physiology , Amyloid Precursor Protein Secretases/chemistry , Amyloid beta-Protein Precursor/chemistry , Animals , Cells, Cultured , Enzyme Activation , Extracellular Space , HEK293 Cells , Humans , Membrane Glycoproteins/chemistry , Mice , Models, Molecular , Molecular Docking Simulation , Protein Binding , Protein Structure, Quaternary , Proteolysis , Structure-Activity Relationship
5.
Bioorg Med Chem Lett ; 29(14): 1737-1745, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31122869

ABSTRACT

The discovery, design and synthesis of a new series of GSMs is described. The classical imidazole heterocycle has been replaced by a cyano group attached to an indole nucleus. The exploration of this series has led to compound 26-S which combined high in vitro and in vivo potency with an acceptable drug-like profile.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Indoles/chemical synthesis , Drug Design , Humans , Structure-Activity Relationship
7.
8.
ACS Med Chem Lett ; 4(4): 419-22, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-24900687

ABSTRACT

The synthesis and preclinical characterization of two novel, brain penetrating P2X7 compounds will be described. Both compounds are shown to be high potency P2X7 antagonists in human, rat, and mouse cell lines and both were shown to have high brain concentrations and robust receptor occupancy in rat. Compound 7 is of particular interest as a probe compound for the preclinical assessment of P2X7 blockade in animal models of neuro-inflammation.

9.
Synapse ; 66(12): 1002-14, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22927303

ABSTRACT

The metabotropic glutamate 1 receptor (mGlu1) is an important protein in the regulation of glutamate transmission in the brain, and believed to be involved in disorders such as ischemia, epilepsy, neuropathic pain, anxiety, and schizophrenia. The goal of this study was to evaluate two selective mGlu1 antagonists [(11) C]3 and [(18) F]4 as potential PET radioligands for the in vivo imaging of the mGlu1 receptor. Biodistribution studies in rats indicated high uptake of [(11) C]3 and [(18) F]4 in the brain. The highest activity level was found in the cerebellum, followed by striatum, hippocampus, frontal cortex, and medulla, in a pattern consistent with the distribution of mGlu1 receptor in rat. At 30 min postinjection, the activity ratio of cerebellum to medulla was 4.5 for [(11) C]3, indicating a high degree of specific binding, while specific binding was lower for [(18) F]4 (cerebellum to medulla activity ratio of 2.0). Moreover, binding of the radioligands [(11) C]3 and [(18) F]4 in mGlu1 receptor-rich region such as cerebellum was blocked by pretreatment of the rats with their respective unlabeled compound or the selective mGlu1 antagonist (compound 5, 2 mg/kg each), but not by the selective mGlu2 antagonist LY341495, or the selective mGlu5 antagonist MPEP (2 mg/kg), thus indicating the binding specificity and selectivity of [(11) C]3 and [(18) F]4 to the mGlu1 receptor. However, in imaging experiments in baboons [(11) C]3 displayed a small specific binding signal only in the cerebellum, while the specific binding of [(18) F]4 was difficult to detect. Species differences in receptor density and affinity of the radioligands in large part account for the differences in the behavior of [(11) C]3 and [(18) F]4 in rats and baboons. Radioligands with higher affinity and/or lower lipophilicity are needed to successfully image the mGlu1 receptor in humans.


Subject(s)
Positron-Emission Tomography , Quinolines/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Receptors, Metabotropic Glutamate/metabolism , Amino Acids/pharmacology , Animals , Brain/diagnostic imaging , CHO Cells , Carbon Radioisotopes/pharmacokinetics , Cricetinae , Cricetulus , Excitatory Amino Acid Antagonists/pharmacology , Fluorine Radioisotopes/pharmacokinetics , Ligands , Male , Papio , Pyridines/pharmacology , Quinolines/chemical synthesis , Quinolines/chemistry , Radiopharmaceuticals/chemical synthesis , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Tissue Distribution , Xanthenes/pharmacology
10.
J Med Chem ; 55(21): 9089-106, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-22650177

ABSTRACT

The design and the synthesis of several chemical subclasses of imidazole containing γ-secretase modulators (GSMs) is described. Conformational restriction of pyridone 4 into bicyclic pyridone isosteres has led to compounds with high in vitro and in vivo potency. This has resulted in the identification of benzimidazole 44a as a GSM with low nanomolar potency in vitro. In mouse, rat, and dog, this compound displayed the typical γ-secretase modulatory profile by lowering Aß42 and Aß40 levels combined with an especially pronounced increase in Aß38 and Aß37 levels while leaving the total levels of amyloid peptides unchanged.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Benzimidazoles/chemical synthesis , Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Imidazoles/chemical synthesis , Amyloid beta-Peptides/metabolism , Animals , Benzimidazoles/pharmacokinetics , Benzimidazoles/pharmacology , Benzoxazoles/chemical synthesis , Benzoxazoles/pharmacokinetics , Benzoxazoles/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Dogs , Drug Design , Humans , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Indazoles/chemical synthesis , Indazoles/pharmacokinetics , Indazoles/pharmacology , Male , Mice , Microsomes, Liver/metabolism , Molecular Conformation , Peptide Fragments/metabolism , Pyridines/chemical synthesis , Pyridines/pharmacokinetics , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley
11.
J Med Chem ; 50(18): 4261-4, 2007 Sep 06.
Article in English | MEDLINE | ID: mdl-17685503

ABSTRACT

A new aspartic protease inhibitory chemotype bearing a 2-amino-3,4-dihydroquinazoline ring was identified by high-throughput screening for the inhibition of BACE-1. X-ray crystallography revealed that the exocyclic amino group participated in a hydrogen bonding array with the two catalytic aspartic acids of BACE-1 (Asp(32), Asp(228)). BACE-1 inhibitory potency was increased (0.9 microM to 11 nM K(i)) by substitution into the unoccupied S(1)' pocket.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Models, Molecular , Quinazolines/chemical synthesis , Amyloid Precursor Protein Secretases/chemistry , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/chemistry , CHO Cells , Caco-2 Cells , Cell Membrane Permeability , Cricetinae , Cricetulus , Crystallography, X-Ray , Humans , Hydrogen Bonding , Molecular Conformation , Mutation , Oligopeptides/chemistry , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/blood , Peptide Fragments/metabolism , Quinazolines/chemistry , Quinazolines/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship
12.
J Med Chem ; 48(16): 5096-9, 2005 Aug 11.
Article in English | MEDLINE | ID: mdl-16078827

ABSTRACT

A selective metabotropic glutamate 1 receptor (mGlu1) antagonist was labeled with the positron-emitting radioisotope carbon-11 and evaluated in ex vivo biodistribution studies and micro-positron emission tomography (micro-PET) imaging experiments in rats. Results from animal experiments demonstrate that the radioligand [11C]2 is the first PET tracer capable of labeling the rat mGlu1 receptor in vivo.


Subject(s)
Quinolines/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Receptors, Metabotropic Glutamate/metabolism , Animals , Brain/diagnostic imaging , Brain/metabolism , Carbon Radioisotopes , Ligands , Positron-Emission Tomography , Quinolines/chemistry , Quinolines/pharmacology , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacology , Rats , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Structure-Activity Relationship , Tissue Distribution
13.
J Med Chem ; 48(6): 2134-53, 2005 Mar 24.
Article in English | MEDLINE | ID: mdl-15771457

ABSTRACT

We describe the discovery and the structure-activity relationship of a new series of quinoline derivatives acting as selective and highly potent noncompetitive mGlu1 antagonists. We first identified cis-10 as a fairly potent mGlu1 antagonist (IC(50) = 20 nM) in a cell-based signal transduction assay on the rat mGlu1 receptor expressed in CHO-K1 cells, and then we were able to design and synthesize highly potent compounds on both rat and human mGlu1 receptors as exemplified by compound cis-64a, which has an antagonist potency of 0.5 nM for the human mGlu1 receptor. We briefly present and discuss the in vitro metabolic stability of the compounds in human liver microsomes. We finally report the pharmacokinetic properties of our lead compound cis-64a.


Subject(s)
Quinolines/chemical synthesis , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Animals , Biological Availability , CHO Cells , Calcium/metabolism , Cell Line, Tumor , Cricetinae , Cricetulus , Half-Life , Humans , In Vitro Techniques , Intracellular Fluid/metabolism , Microsomes, Liver/metabolism , Quinolines/pharmacokinetics , Quinolines/pharmacology , Rats , Receptors, Metabotropic Glutamate/physiology , Signal Transduction/drug effects , Stereoisomerism , Structure-Activity Relationship
14.
Neuropharmacology ; 47(7): 961-72, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15555631

ABSTRACT

We examined the pharmacological profile of (3,4-dihydro-2H-pyrano[2,3]b quinolin-7-yl) (cis-4-methoxycyclohexyl) methanone (JNJ16259685). At recombinant rat and human metabotropic glutamate (mGlu) 1a receptors, JNJ16259685 non-competitively inhibited glutamate-induced Ca2+ mobilization with IC50 values of 3.24+/-1.00 and 1.21+/-0.53 nM, respectively, while showing a much lower potency at the rat and human mGlu5a receptor. JNJ16259685 inhibited [3H]1-(3,4-dihydro-2H-pyrano[2,3-b]quinolin-7-yl)-2-phenyl-1-ethanone ([3H]R214127) binding to membranes prepared from cells expressing rat mGlu1a receptors with a Ki of 0.34+/-0.20 nM. JNJ16259685 showed no agonist, antagonist or positive allosteric activity toward rat mGlu2, -3, -4 or -6 receptors at concentrations up to 10 microM and did not bind to AMPA or NMDA receptors, or to a battery of other neurotransmitter receptors, ion channels and transporters. In primary cerebellar cultures, JNJ16259685 inhibited glutamate-mediated inositol phosphate production with an IC50 of 1.73+/-0.40 nM. Subcutaneously administered JNJ16259685 exhibited high potencies in occupying central mGlu1 receptors in the rat cerebellum and thalamus ( ED50=0.040 and 0.014 mg/kg, respectively). These data show that JNJ16259685 is a selective mGlu1 receptor antagonist with excellent potencies in inhibiting mGlu1 receptor function and binding and in occupying the mGlu1 receptor after systemic administration.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Quinolines/pharmacology , Receptors, AMPA/antagonists & inhibitors , Animals , Autoradiography , CHO Cells , Cells, Cultured , Cerebellum/cytology , Cerebellum/drug effects , Cricetinae , Dose-Response Relationship, Drug , Female , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Humans , Inositol Phosphates/metabolism , Male , Pyrans/pharmacology , Rats , Rats, Wistar , Receptors, AMPA/genetics , Recombinant Proteins/drug effects , Tetrahydrofolate Dehydrogenase/deficiency , Transfection
15.
Mol Pharmacol ; 63(5): 1082-93, 2003 May.
Article in English | MEDLINE | ID: mdl-12695537

ABSTRACT

R214127 was shown to be a potent and noncompetitive metabotropic glutamate 1 (mGlu1) receptor-selective antagonist. The kinetics and pharmacology of [(3)H]1-(3,4-dihydro-2H-pyrano[2,3-b]quinolin-7-yl)-2-phenyl-1-ethanone (R214127) binding to rat mGlu1a receptor Chinese hamster ovary (CHO)-dhfr(-) membranes was investigated, as well as the distribution of [(3)H]R214127 binding in rat brain tissue and sections. Specific binding to rat mGlu1a receptor CHO-dhfr(-) membranes was approximately 92% of total and was optimal at 4 degrees C. Full association was reached within 5 min, and [(3)H]R214127 bound to a single binding site with an apparent K(D) of 0.90 +/- 0.14 nM and a B(max) of 6512 +/- 1501 fmol/mg of protein. Inhibition experiments showed that [(3)H]R214127 binding was completely blocked by 2-quinoxaline-carboxamide-N-adamantan-1-yl (NPS 2390), (3aS,6aS)-6a-naphtalan-2-ylmethyl-5-methyliden-hexahydro-cyclopenta[c]furan-1-on (BAY 36-7620), and 7-(hydroxyimino)cyclo-propa[b]chromen-1a-carboxylate ethyl ester (CPCCOEt), but was not displaced by competitive mGlu1 receptor ligands such as glutamate and quisqualate, suggesting that R214127, NPS 2390, BAY 36-7620, and CPCCOEt bind to the same site or mutually exclusive sites. Experiments using rat cortex, striatum, hippocampus and cerebellum revealed that [(3)H]R214127 labeled a single high-affinity binding site (K(D) approximately 1 nM). B(max) values were highest in the cerebellum (4302 +/- 2042 fmol/mg of protein) and were 741 +/- 48, 688 +/- 125, and 471 +/- 68 fmol/mg of protein in the striatum, hippocampus, and cortex, respectively. The distribution of [(3)H]R214127 binding in rat brain was investigated in more detail by radioligand autoradiography. A high density of binding sites was detected in the molecular layer of the cerebellum. Moderate labeling was seen in the CA3 and dentate gyrus of the hippocampus, thalamus, olfactory tubercle, amygdala, and substantia nigra reticulata. The cerebral cortex, caudate putamen, ventral pallidum, and nucleus accumbens showed lower labeling. The high affinity and selectivity of [(3)H]R214127 for mGlu1 receptors renders this compound the ligand of choice to study the native mGlu1 receptor in brain.


Subject(s)
Adamantane/analogs & derivatives , Pyrans/pharmacology , Quinolines/pharmacology , Quinoxalines/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Adamantane/pharmacology , Allosteric Regulation , Animals , Binding Sites , Binding, Competitive , Brain/drug effects , Brain/metabolism , CHO Cells , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Chromones/pharmacology , Cricetinae , Humans , Naphthalenes/pharmacology , Quisqualic Acid/pharmacology , Radioligand Assay , Rats , Transfection , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL
...