Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Bioorg Med Chem Lett ; 30(20): 127471, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32781219

ABSTRACT

Ubiquitin specific protease-7 (USP7) is considered an attractive target for cancer therapy by promoting degradation of the tumor suppressor p53 and negatively affecting the immune response to tumors. However, the development of selective non-covalent USP7 inhibitors has proven challenging. In this work we report the NMR characterization of a weak binder from SPR screening of an in-house fragment library which reveals that it binds to the allosteric palm site of the catalytic domain. Molecular modeling combined with 1HNMR saturation transfer difference and NOESY experiments enabled structure-based design of additional compounds showing IC50 values in the low-micromolar range with good selectivity over the closest homolog USP47. The most potent analogue represents a promising starting point for the development of novel, selective USP7 inhibitors.


Subject(s)
Amides/pharmacology , Drug Discovery , Small Molecule Libraries/pharmacology , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , Allosteric Site/drug effects , Amides/chemical synthesis , Amides/chemistry , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Ubiquitin-Specific Peptidase 7/metabolism
2.
J Neurosci ; 33(24): 10075-84, 2013 Jun 12.
Article in English | MEDLINE | ID: mdl-23761903

ABSTRACT

Aß, the product of APP (amyloid precursor protein), has been implicated in the pathophysiology of Alzheimer's disease (AD). ß-Site APP cleaving enzyme1 (BACE1) is the enzyme initiating the processing of the APP to Aß peptides. Small molecule BACE1 inhibitors are expected to decrease Aß-peptide generation and thereby reduce amyloid plaque formation in the brain, a neuropathological hallmark of AD. BACE1 inhibition thus addresses a key mechanism in AD and its potential as a therapeutic target is currently being addressed in clinical studies. Here, we report the discovery and the pharmacokinetic and pharmacodynamic properties of BACE1 inhibitor AZ-4217, a high potency compound (IC50 160 pM in human SH-SY5Y cells) with an excellent in vivo efficacy. Central efficacy of BACE1 inhibition was observed after a single dose in C57BL/6 mice, guinea pigs, and in an APP transgenic mouse model of cerebral amyloidosis (Tg2576). Furthermore, we demonstrate that in a 1 month treatment paradigm BACE1 inhibition of Aß production does lower amyloid deposition in 12-month-old Tg2576 mice. These results strongly support BACE1 inhibition as concretely impacting amyloid deposition and therefore potentially an important approach for therapeutic intervention in AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid/metabolism , Enzyme Inhibitors/pharmacology , Neurons/drug effects , Neurons/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/metabolism , Cells, Cultured , Cerebral Cortex/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Embryo, Mammalian , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Female , Guinea Pigs , Humans , Isoindoles/pharmacology , Isoindoles/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Peptide Fragments/metabolism , Pyridones/pharmacology , Pyridones/therapeutic use , Time Factors
3.
J Biol Chem ; 287(49): 41245-57, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23048024

ABSTRACT

ß-Site amyloid precursor protein cleaving enzyme1 (BACE1) is one of the key enzymes involved in the processing of the amyloid precursor protein (APP) and formation of amyloid ß peptide (Aß) species. Because cerebral deposition of Aß species might be critical for the pathogenesis of Alzheimer disease, BACE1 has emerged as a key target for the treatment of this disease. Here, we report the discovery and comprehensive preclinical characterization of AZD3839, a potent and selective inhibitor of human BACE1. AZD3839 was identified using fragment-based screening and structure-based design. In a concentration-dependent manner, AZD3839 inhibited BACE1 activity in a biochemical fluorescence resonance energy transfer (FRET) assay, Aß and sAPPß release from modified and wild-type human SH-SY5Y cells and mouse N2A cells as well as from mouse and guinea pig primary cortical neurons. Selectivity against BACE2 and cathepsin D was 14 and >1000-fold, respectively. AZD3839 exhibited dose- and time-dependent lowering of plasma, brain, and cerebrospinal fluid Aß levels in mouse, guinea pig, and non-human primate. Pharmacokinetic/pharmacodynamic analyses of mouse and guinea pig data showed a good correlation between the potency of AZD3839 in primary cortical neurons and in vivo brain effects. These results suggest that AZD3839 effectively reduces the levels of Aß in brain, CSF, and plasma in several preclinical species. It might, therefore, have disease-modifying potential in the treatment of Alzheimer disease and related dementias. Based on the overall pharmacological profile and its drug like properties, AZD3839 has been progressed into Phase 1 clinical trials in man.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/cerebrospinal fluid , Aspartic Acid Endopeptidases/antagonists & inhibitors , Indoles/pharmacology , Pyrimidines/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Biomarkers/metabolism , Brain/metabolism , Cathepsin D/metabolism , Cell Line , Disease Progression , Dose-Response Relationship, Drug , Drug Design , Drug Evaluation, Preclinical , Female , Fluorescence Resonance Energy Transfer/methods , Guinea Pigs , Humans , Male , Mice , Mice, Inbred C57BL , Treatment Outcome
4.
Drug Metab Dispos ; 41(5): 1134-47, 2013 May.
Article in English | MEDLINE | ID: mdl-23474650

ABSTRACT

Recently, the discovery of the aminoisoindoles as potent and selective inhibitors of ß-secretase was reported, including the close structural analogs compound (S)-1-pyridin-4-yl-4-fluoro-1-(3-(pyrimidin-5-yl)phenyl)-1H-isoindol-3-amine [(S)-25] and (S)-1-(2-(difluoromethyl)pyridin-4-yl)-4-fluoro-1-(3-(pyrimidin-5-yl)phenyl)-1H-isoindol-3-amine hemifumarate (AZD3839), the latter being recently progressed to the clinic. The biotransformation of (S)-25 was investigated in vitro and in vivo in rat, rabbit, and human and compared with AZD3839 to further understand the metabolic fate of these compounds. In vitro, CYP3A4 was the major responsible enzyme and metabolized both compounds to a large extent in the commonly shared pyridine and pyrimidine rings. The main proposed metabolic pathways in various in vitro systems were N-oxidation of the pyridine and/or pyrimidine ring and conversion to 4-pyrimidone and pyrimidine-2,4-dione. Both compounds were extensively metabolized, and more than 90% was excreted in feces after intravenous administration of radiolabeled compound to the rat. Here, the main pathways were N-oxidation of the pyridine and/or pyrimidine ring and a ring contraction of the pyrimidine ring into an imidazole ring. Ring-contracted metabolites accounted for 25% of the total metabolism in the rat for (S)-25, whereas the contribution was much smaller for AZD3839. This metabolic pathway was not foreseen on the basis of the obtained in vitro data. In conclusion, we discovered an unusual metabolic pathway of aryl-pyrimidine-containing compounds by a ring-opening reaction followed by elimination of a carbon atom and a ring closure to form an imidazole ring.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Pyrimidines/chemistry , Animals , Biotransformation , Chromatography, Liquid , Cyclization , Enzyme Inhibitors/chemistry , Humans , Indoles/pharmacokinetics , Male , Mass Spectrometry , Pyrimidines/pharmacokinetics , Rabbits , Rats , Rats, Sprague-Dawley
5.
Org Lett ; 5(9): 1451-4, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12713296

ABSTRACT

A new palladium-dipyridylmethylamine complex is an excellent catalyst for C-C bond-forming processes such as the Heck, Suzuki, and Sonogashira reactions in organic and aqueous solvents under homogeneous conditions. [reaction: see text]

6.
J Med Chem ; 56(8): 3177-90, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23516963

ABSTRACT

We have developed two parallel series, A and B, of CX3CR1 antagonists for the treatment of multiple sclerosis. By modifying the substituents on the 7-amino-5-thio-thiazolo[4,5-d]pyrimidine core structure, we were able to achieve compounds with high selectivity for CX3CR1 over the closely related CXCR2 receptor. The structure-activity relationships showed that a leucinol moiety attached to the core-structure in the 7-position together with α-methyl branched benzyl derivatives in the 5-position displayed promising affinity, and selectivity as well as physicochemical properties, as exemplified by compounds 18a and 24h. We show the preparation of the first potent and selective orally available CX3CR1 antagonists.


Subject(s)
Multiple Sclerosis/drug therapy , Pyrimidines/pharmacology , Receptors, Chemokine/antagonists & inhibitors , Thiazoles/pharmacology , Amino Alcohols/chemical synthesis , Amino Alcohols/pharmacokinetics , Amino Alcohols/pharmacology , Animals , CX3C Chemokine Receptor 1 , Caco-2 Cells , Humans , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Rats , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry , Thiazoles/pharmacokinetics
7.
J Med Chem ; 55(21): 9297-311, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-23017051

ABSTRACT

Amino-2H-imidazoles are described as a new class of BACE-1 inhibitors for the treatment of Alzheimer's disease. Synthetic methods, crystal structures, and structure-activity relationships for target activity, permeability, and hERG activity are reported and discussed. Compound (S)-1m was one of the most promising compounds in this report, with high potency in the cellular assay and a good overall profile. When guinea pigs were treated with compound (S)-1m, a concentration and time dependent decrease in Aß40 and Aß42 levels in plasma, brain, and CSF was observed. The maximum reduction of brain Aß was 40-50%, 1.5 h after oral dosing (100 µmol/kg). The results presented highlight the potential of this new class of BACE-1 inhibitors with good target potency and with low effect on hERG, in combination with a fair CNS exposure in vivo.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/antagonists & inhibitors , Brain/drug effects , Imidazoles/chemical synthesis , Peptide Fragments/metabolism , Amyloid Precursor Protein Secretases/chemistry , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/cerebrospinal fluid , Animals , Aspartic Acid Endopeptidases/chemistry , Brain/metabolism , Cell Line , Crystallography, X-Ray , Dogs , Female , Guinea Pigs , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Peptide Fragments/blood , Peptide Fragments/cerebrospinal fluid , Permeability , Stereoisomerism , Structure-Activity Relationship , Tissue Distribution
8.
J Med Chem ; 55(21): 9346-61, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-22924815

ABSTRACT

The evaluation of a series of aminoisoindoles as ß-site amyloid precursor protein cleaving enzyme 1 (BACE1) inhibitors and the discovery of a clinical candidate drug for Alzheimer's disease, (S)-32 (AZD3839), are described. The improvement in permeability properties by the introduction of fluorine adjacent to the amidine moiety, resulting in in vivo brain reduction of Aß40, is discussed. Due to the basic nature of these compounds, they displayed affinity for the human ether-a-go-go related gene (hERG) ion channel. Different ways to reduce hERG inhibition and increase hERG margins for this series are described, culminating in (S)-16 and (R)-41 showing large in vitro margins with BACE1 cell IC(50) values of 8.6 and 0.16 nM, respectively, and hERG IC(50) values of 16 and 2.8 µM, respectively. Several compounds were advanced into pharmacodynamic studies and demonstrated significant reduction of ß-amyloid peptides in mouse brain following oral dosing.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Indoles/chemical synthesis , Pyrimidines/chemical synthesis , Administration, Oral , Alkynes/chemical synthesis , Alkynes/pharmacokinetics , Alkynes/pharmacology , Amides/chemical synthesis , Amides/pharmacokinetics , Amides/pharmacology , Amyloid Precursor Protein Secretases/chemistry , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/chemistry , Biological Availability , Brain/drug effects , Brain/metabolism , Cell Line , Crystallography, X-Ray , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Female , Fluorescence Resonance Energy Transfer , Humans , Hydrogen Bonding , Indoles/pharmacokinetics , Indoles/pharmacology , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Peptide Fragments/metabolism , Permeability , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Stereoisomerism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL