Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Mol Ther Nucleic Acids ; 29: 625-642, 2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36090761

ABSTRACT

Tau is a microtubule-associated protein (MAPT, tau) implicated in the pathogenesis of tauopathies, a spectrum of neurodegenerative disorders characterized by accumulation of hyperphosphorylated, aggregated tau. Because tau pathology can be distinct across diseases, a pragmatic therapeutic approach may be to intervene at the level of the tau transcript, as it makes no assumptions to mechanisms of tau toxicity. Here we performed a large library screen of locked-nucleic-acid (LNA)-modified antisense oligonucleotides (ASOs), where careful tiling of the MAPT locus resulted in the identification of hot spots for activity in the 3' UTR. Further modifications to the LNA design resulted in the generation of ASO-001933, which selectively and potently reduces tau in primary cultures from hTau mice, monkey, and human neurons. ASO-001933 was well tolerated and produced a robust, long-lasting reduction in tau protein in both mouse and cynomolgus monkey brain. In monkey, tau protein reduction was maintained in brain for 20 weeks post injection and corresponded with tau protein reduction in the cerebrospinal fluid (CSF). Our results demonstrate that LNA-ASOs exhibit excellent drug-like properties and sustained efficacy likely translating to infrequent, intrathecal dosing in patients. These data further support the development of LNA-ASOs against tau for the treatment of tauopathies.

2.
ACS Med Chem Lett ; 10(3): 312-317, 2019 Mar 14.
Article in English | MEDLINE | ID: mdl-30891132

ABSTRACT

A triazine hit identified from a screen of the BMS compound collection was optimized for potency, in vivo activity, and off-target profile to produce the bicyclic pyrimidine γ-secretase modulator BMS-932481. The compound showed robust reductions of Aß1-42 and Aß1-40 in the plasma, brain, and cerebrospinal fluid of mice and rats. Consistent with the γ-secretase modulator mechanism, increases in Aß1-37 and Aß1-38 were observed, with no change in the total amount of Aß1-x produced. No Notch-based toxicity was observed, and the overall preclinical profile of BMS-932481 supported its further evaluation in human clinical trials.

3.
Eur J Pharmacol ; 812: 104-112, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28690193

ABSTRACT

Alzheimer's disease is associated with the accumulation of amyloid-ß (Aß) in the brain. In particular, the 42-amino acid form, Aß1-42, is thought to play a key role in the disease. It is therefore of interest that diverse compounds, known as γ-secretase modulators (GSM), can selectively decrease Aß1-42 production without inhibiting the production of other forms of Aß. Here we describe the novel discovery of synergistic inhibition of Aß by certain combinations of GSMs. Cell cultures were treated with pairwise combinations of GSMs to determine how Aß peptide production was affected. Analysis of isobolograms and calculation of the combination index showed that BMS-869780 and GSM-2 were highly synergistic. Additional combinations of GSMs revealed that inhibition of Aß occurred only when one GSM was of the "acid GSM" structural class and the other was of the "non-acid GSM" class. A total of 15 representative acid/non-acid GSM combinations were shown to inhibit Aß production, whereas 10 pairwise combinations containing two acid GSMs or containing two non-acid GSMs did not inhibit Aß. We also discovered that lasalocid, a natural product, is a potent GSM. Lasalocid is unique in that it did not synergize with other GSMs. Synergism did not translate in vivo perhaps because of biochemical differences between the cell culture model and brain. These findings reinforce the pharmacological differences between different structural classes of GSMs, and may help to exploit the potential of γ-secretase as a drug target.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/biosynthesis , Peptide Fragments/biosynthesis , Protease Inhibitors/pharmacology , Acetates/pharmacology , Animals , Cell Line, Tumor , Drug Synergism , Humans , Mice , Piperidines/pharmacology
4.
J Pharmacol Exp Ther ; 358(1): 125-37, 2016 07.
Article in English | MEDLINE | ID: mdl-27189974

ABSTRACT

The amyloid-ß peptide (Aß)-in particular, the 42-amino acid form, Aß1-42-is thought to play a key role in the pathogenesis of Alzheimer's disease (AD). Thus, several therapeutic modalities aiming to inhibit Aß synthesis or increase the clearance of Aß have entered clinical trials, including γ-secretase inhibitors, anti-Aß antibodies, and amyloid-ß precursor protein cleaving enzyme inhibitors. A unique class of small molecules, γ-secretase modulators (GSMs), selectively reduce Aß1-42 production, and may also decrease Aß1-40 while simultaneously increasing one or more shorter Aß peptides, such as Aß1-38 and Aß1-37. GSMs are particularly attractive because they do not alter the total amount of Aß peptides produced by γ-secretase activity; they spare the processing of other γ-secretase substrates, such as Notch; and they do not cause accumulation of the potentially toxic processing intermediate, ß-C-terminal fragment. This report describes the translation of pharmacological activity across species for two novel GSMs, (S)-7-(4-fluorophenyl)-N2-(3-methoxy-4-(3-methyl-1H-1,2,4-triazol-1-yl)phenyl)-N4-methyl-6,7-dihydro-5H-cyclopenta[d]pyrimidine-2,4-diamine (BMS-932481) and (S,Z)-17-(4-chloro-2-fluorophenyl)-34-(3-methyl-1H-1,2,4-triazol-1-yl)-16,17-dihydro-15H-4-oxa-2,9-diaza-1(2,4)-cyclopenta[d]pyrimidina-3(1,3)-benzenacyclononaphan-6-ene (BMS-986133). These GSMs are highly potent in vitro, exhibit dose- and time-dependent activity in vivo, and have consistent levels of pharmacological effect across rats, dogs, monkeys, and human subjects. In rats, the two GSMs exhibit similar pharmacokinetics/pharmacodynamics between the brain and cerebrospinal fluid. In all species, GSM treatment decreased Aß1-42 and Aß1-40 levels while increasing Aß1-38 and Aß1-37 by a corresponding amount. Thus, the GSM mechanism and central activity translate across preclinical species and humans, thereby validating this therapeutic modality for potential utility in AD.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Aniline Compounds/pharmacology , Aniline Compounds/pharmacokinetics , Brain/drug effects , Bridged-Ring Compounds/pharmacology , Bridged-Ring Compounds/pharmacokinetics , Pyrimidines/pharmacology , Pyrimidines/pharmacokinetics , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/genetics , Aniline Compounds/chemistry , Animals , Brain/enzymology , Brain/metabolism , Bridged-Ring Compounds/chemistry , Cell Line , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Humans , Macaca fascicularis , Pyrimidines/chemistry , Rats, Sprague-Dawley , Receptors, Notch/metabolism , Species Specificity , Tissue Distribution
5.
Bioorg Med Chem Lett ; 25(22): 5040-7, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26497283

ABSTRACT

The synthesis, evaluation, and structure-activity relationships of a class of acyl guanidines which inhibit the BACE-1 enzyme are presented. The prolinyl acyl guanidine chemotype (7c), unlike compounds of the parent isothiazole chemotype (1), yielded compounds with good agreement between their enzymatic and cellular potency as well as a reduced susceptibility to P-gp efflux. Further improvements in potency and P-gp ratio were realized via a macrocyclization strategy. The in vivo profile in wild-type mice and P-gp effects for the macrocyclic analog 21c is presented.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Guanidines/pharmacology , Macrocyclic Compounds/pharmacology , Proline/analogs & derivatives , Proline/pharmacology , Protease Inhibitors/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Amyloid beta-Peptides/biosynthesis , Animals , Caco-2 Cells , Cathepsin D/antagonists & inhibitors , Cathepsin E/antagonists & inhibitors , Dogs , Guanidines/chemical synthesis , Humans , Macrocyclic Compounds/chemical synthesis , Madin Darby Canine Kidney Cells , Male , Mice , Molecular Docking Simulation , Pepsin A/antagonists & inhibitors , Proline/chemical synthesis , Protease Inhibitors/chemical synthesis
6.
Anal Biochem ; 466: 65-71, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25175011

ABSTRACT

Microtubules (MTs) are highly dynamic polymers composed of α- and ß-tubulin heterodimers. Dysregulation of MT dynamics in neurons may be a contributing factor in the progression of various neurodegenerative diseases. We developed a stable isotope labeling by amino acids in cell culture (SILAC)-based liquid chromatography-mass spectrometry (LC-MS) method to measure the fraction of [(13)C6]leucine-labeled α-tubulin-derived surrogate peptides. Using this approach, we measured the time course of incorporation of [(13)C6]leucine label into the MT and dimer pools isolated from cycling cells and rat primary hippocampal neurons. We found that the MT pool is in rapid equilibrium with the dimer pool in the cycling cells, consistent with rapid MT polymerization/depolymerization during cell proliferation. Conversely, in neurons, we found that labeling of the MT pool was rapid, whereas the dimer pool was delayed. These results suggest that newly synthesized α-tubulin is first incorporated into MTs or complexes that co-sediment with MTs and that appearance of labeled α-tubulin in the dimer pool may be a consequence of MT depolymerization or breakdown. Our results demonstrate that a SILAC-based approach can be used to measure MT dynamics and may have utility for exploring MT dysregulation in various models of neurodegenerative disease.


Subject(s)
Biological Assay/methods , Isotope Labeling/methods , Microtubules/metabolism , Neurons/metabolism , Animals , Brain/cytology , Cell Culture Techniques , Chromatography, Liquid , Mass Spectrometry , Neurons/cytology , Peptides/chemistry , Rats , Reproducibility of Results
7.
Int J Alzheimers Dis ; 2014: 431858, 2014.
Article in English | MEDLINE | ID: mdl-25097793

ABSTRACT

Alzheimer's disease is the most prevalent cause of dementia and is associated with accumulation of amyloid-ß peptide (Aß), particularly the 42-amino acid Aß1-42, in the brain. Aß1-42 levels can be decreased by γ-secretase modulators (GSM), which are small molecules that modulate γ-secretase, an enzyme essential for Aß production. BMS-869780 is a potent GSM that decreased Aß1-42 and Aß1-40 and increased Aß1-37 and Aß1-38, without inhibiting overall levels of Aß peptides or other APP processing intermediates. BMS-869780 also did not inhibit Notch processing by γ-secretase and lowered brain Aß1-42 without evidence of Notch-related side effects in rats. Human pharmacokinetic (PK) parameters were predicted through allometric scaling of PK in rat, dog, and monkey and were combined with the rat pharmacodynamic (PD) parameters to predict the relationship between BMS-869780 dose, exposure and Aß1-42 levels in human. Off-target and safety margins were then based on comparisons to the predicted exposure required for robust Aß1-42 lowering. Because of insufficient safety predictions and the relatively high predicted human daily dose of 700 mg, further evaluation of BMS-869780 as a potential clinical candidate was discontinued. Nevertheless, BMS-869780 demonstrates the potential of the GSM approach for robust lowering of brain Aß1-42 without Notch-related side effects.

8.
PLoS One ; 8(10): e76523, 2013.
Article in English | MEDLINE | ID: mdl-24116116

ABSTRACT

Cerebral spinal fluid (CSF) Aß42, tau and p181tau are widely accepted biomarkers of Alzheimer's disease (AD). Numerous studies show that CSF tau and p181tau levels are elevated in mild-to-moderate AD compared to age-matched controls. In addition, these increases might predict preclinical AD in cognitively normal elderly. Despite their importance as biomarkers, the molecular nature of CSF tau and ptau is not known. In the current study, reverse-phase high performance liquid chromatography was used to enrich and concentrate tau prior to western-blot analysis. Multiple N-terminal and mid-domain fragments of tau were detected in pooled CSF with apparent sizes ranging from <20 kDa to ~40 kDa. The pattern of tau fragments in AD and control samples were similar. In contrast, full-length tau and C-terminal-containing fragments were not detected. To quantify levels, five tau ELISAs and three ptau ELISAs were developed to detect different overlapping regions of the protein. The discriminatory potential of each assay was determined using 20 AD and 20 age-matched control CSF samples. Of the tau ELISAs, the two assays specific for tau containing N-terminal sequences, amino acids 9-198 (numbering based on tau 441) and 9-163, exhibited the most significant differences between AD and control samples. In contrast, CSF tau was not detected with an ELISA specific for a more C-terminal region (amino acids 159-335). Significant discrimination was also observed with ptau assays measuring amino acids 159-p181 and 159-p231. Interestingly, the discriminatory potential of p181 was reduced when measured in the context of tau species containing amino acids 9-p181. Taken together, these results demonstrate that tau in CSF occurs as a series of fragments and that discrimination of AD from control is dependent on the subset of tau species measured. These assays provide novel tools to investigate CSF tau and ptau as biomarkers for other neurodegenerative diseases.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Peptide Fragments/cerebrospinal fluid , tau Proteins/cerebrospinal fluid , Aged , Aged, 80 and over , Alzheimer Disease/diagnosis , Blotting, Western , Chromatography, High Pressure Liquid , Chromatography, Reverse-Phase , Enzyme-Linked Immunosorbent Assay/methods , Humans , Male , Middle Aged , Molecular Weight , Peptide Fragments/metabolism , Phosphorylation , Reproducibility of Results , Sensitivity and Specificity , tau Proteins/chemistry , tau Proteins/metabolism
9.
J Pharmacol Exp Ther ; 344(3): 686-95, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23275065

ABSTRACT

A hallmark of Alzheimer's disease (AD) pathology is the accumulation of brain amyloid ß-peptide (Aß), generated by γ-secretase-mediated cleavage of the amyloid precursor protein (APP). Therefore, γ-secretase inhibitors (GSIs) may lower brain Aß and offer a potential new approach to treat AD. As γ-secretase also cleaves Notch proteins, GSIs can have undesirable effects due to interference with Notch signaling. Avagacestat (BMS-708163) is a GSI developed for selective inhibition of APP over Notch cleavage. Avagacestat inhibition of APP and Notch cleavage was evaluated in cell culture by measuring levels of Aß and human Notch proteins. In rats, dogs, and humans, selectivity was evaluated by measuring plasma blood concentrations in relation to effects on cerebrospinal fluid (CSF) Aß levels and Notch-related toxicities. Measurements of Notch-related toxicity included goblet cell metaplasia in the gut, marginal-zone depletion in the spleen, reductions in B cells, and changes in expression of the Notch-regulated hairy and enhancer of split homolog-1 from blood cells. In rats and dogs, acute administration of avagacestat robustly reduced CSF Aß40 and Aß42 levels similarly. Chronic administration in rats and dogs, and 28-day, single- and multiple-ascending-dose administration in healthy human subjects caused similar exposure-dependent reductions in CSF Aß40. Consistent with the 137-fold selectivity measured in cell culture, we identified doses of avagacestat that reduce CSF Aß levels without causing Notch-related toxicities. Our results demonstrate the selectivity of avagacestat for APP over Notch cleavage, supporting further evaluation of avagacestat for AD therapy.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Protein Precursor/antagonists & inhibitors , Oxadiazoles/pharmacology , Sulfonamides/pharmacology , Adolescent , Adult , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Cells, Cultured , Dogs , Female , Humans , Male , Middle Aged , Rats , Rats, Sprague-Dawley , Receptors, Notch/metabolism , Signal Transduction/drug effects , Young Adult
10.
J Med Chem ; 55(21): 9208-23, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-23030502

ABSTRACT

This report describes the discovery and optimization of a BACE-1 inhibitor series containing an unusual acyl guanidine chemotype that was originally synthesized as part of a 6041-membered solid-phase library. The synthesis of multiple follow-up solid- and solution-phase libraries facilitated the optimization of the original micromolar hit into a single-digit nanomolar BACE-1 inhibitor in both radioligand binding and cell-based functional assay formats. The X-ray structure of representative inhibitors bound to BACE-1 revealed a number of key ligand:protein interactions, including a hydrogen bond between the side chain amide of flap residue Gln73 and the acyl guanidine carbonyl group, and a cation-π interaction between Arg235 and the isothiazole 4-methoxyphenyl substituent. Following subcutaneous administration in rats, an acyl guanidine inhibitor with single-digit nanomolar activity in cells afforded good plasma exposures and a dose-dependent reduction in plasma Aß levels, but poor brain exposure was observed (likely due to Pgp-mediated efflux), and significant reductions in brain Aß levels were not obtained.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Guanidines/chemical synthesis , Small Molecule Libraries , Amyloid Precursor Protein Secretases/chemistry , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/chemistry , Brain/metabolism , Cell Line , Crystallography, X-Ray , Guanidines/pharmacokinetics , Guanidines/pharmacology , Humans , Isoxazoles/chemical synthesis , Isoxazoles/pharmacokinetics , Isoxazoles/pharmacology , Models, Molecular , Molecular Structure , Mutation , Peptide Fragments/metabolism , Protein Binding , Radioligand Assay , Rats , Solid-Phase Synthesis Techniques , Solutions , Structure-Activity Relationship
11.
J Neurosci ; 32(21): 7137-45, 2012 May 23.
Article in English | MEDLINE | ID: mdl-22623658

ABSTRACT

Tau is a microtubule (MT)-stabilizing protein that is altered in Alzheimer's disease (AD) and other tauopathies. It is hypothesized that the hyperphosphorylated, conformationally altered, and multimeric forms of tau lead to a disruption of MT stability; however, direct evidence is lacking in vivo. In this study, an in vivo stable isotope-mass spectrometric technique was used to measure the turnover, or dynamicity, of MTs in brains of living animals. We demonstrated an age-dependent increase in MT dynamics in two different tau transgenic mouse models, 3xTg and rTg4510. MT hyperdynamicity was dependent on tau expression, since a reduction of transgene expression with doxycycline reversed the MT changes. Treatment of rTg4510 mice with the epothilone, BMS-241027, also restored MT dynamics to baseline levels. In addition, MT stabilization with BMS-241027 had beneficial effects on Morris water maze deficits, tau pathology, and neurodegeneration. Interestingly, pathological and functional benefits of BMS-241027 were observed at doses that only partially reversed MT hyperdynamicity. Together, these data suggest that tau-mediated loss of MT stability may contribute to disease progression and that very low doses of BMS-241027 may be useful in the treatment of AD and other tauopathies.


Subject(s)
Cognition Disorders/drug therapy , Epothilones/therapeutic use , Microtubules/pathology , Nerve Degeneration/drug therapy , Tauopathies/drug therapy , Tubulin Modulators/therapeutic use , tau Proteins/physiology , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cognition Disorders/complications , Cognition Disorders/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Doxycycline/pharmacology , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/psychology , Epothilones/pharmacology , Female , Hippocampus/drug effects , Hippocampus/pathology , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubules/drug effects , Tauopathies/complications , Tauopathies/genetics , Tauopathies/pathology , Tauopathies/psychology , Tubulin Modulators/pharmacology , tau Proteins/antagonists & inhibitors , tau Proteins/biosynthesis , tau Proteins/genetics
12.
Bioorg Med Chem Lett ; 21(22): 6909-15, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21974952
13.
Bioorg Med Chem Lett ; 21(22): 6916-24, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21782431

ABSTRACT

The synthesis, evaluation, and structure-activity relationships of a class of γ-lactam 1,3-diaminopropan-2-ol transition-state isostere inhibitors of BACE are discussed. Two strategies for optimizing lead compound 1a are presented. Reducing the overall size of the inhibitors resulted in the identification of γ-lactam 1i, whereas the introduction of conformational constraint on the prime-side of the inhibitor generated compounds such as the 3-hydroxypyrrolidine inhibitor 28n. The full in vivo profile of 1i in rats and 28n in Tg 2576 mice is presented.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Lactams/chemistry , Lactams/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Animals , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Lactams/chemical synthesis , Lactams/pharmacokinetics , Mice , Models, Molecular , Rats , Structure-Activity Relationship
14.
J Alzheimers Dis ; 24 Suppl 2: 127-41, 2011.
Article in English | MEDLINE | ID: mdl-21422517

ABSTRACT

Levels of tau in cerebrospinal fluid (CSF) are elevated in Alzheimer's disease (AD) patients. It is believed this elevation is related to the tau pathology and neurodegeneration observed in AD, but not all tauopathies have increased CSF tau. There has been little pre-clinical work to investigate mechanisms of increased CSF tau due to the difficulty in collecting CSF samples from mice, the most commonly used pre-clinical models. We developed methods to collect CSF from mice without contamination from tau in brain tissue, which is approximately 50,000 fold more abundant in brain than CSF. Using these methods, we measured CSF tau from 3xTg, Tg4510, and Tau Alone transgenic mice. All three lines of mice showed age-dependent increases in CSF tau. They varied in phenotype from undetectable to severe tau pathology and neurodegeneration, suggesting that degenerating neurons are unlikely to be the only source of pathologic CSF tau. Overall, CSF tau levels mirrored expression levels and changes of tau in the brain, but they did not always correlate exactly. CSF tau was often more sensitive to changes in brain transgene expression and pathology. In addition, we also developed ELISA assays specific to different regions of the tau protein. We used these assays to provide evidence that CSF tau exists as fragments, with little intact C-terminus and partial loss of the N-terminus. Taken together, these assays and mouse models may be used to facilitate a deeper understanding of CSF tau in neurodegenerative disease.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , tau Proteins/cerebrospinal fluid , Age Factors , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Biomarkers/cerebrospinal fluid , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Doxycycline/pharmacology , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurofilament Proteins/metabolism , Presenilin-1/genetics , RNA, Messenger/metabolism , Tubulin/metabolism , tau Proteins/genetics
15.
Bioorg Med Chem Lett ; 21(1): 537-41, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21078556

ABSTRACT

Heterocyclic replacement of the isophthalamide phenyl ring in hydroxyethylamine (HEA) BACE-1 inhibitors was explored. A variety of indole-1,3-dicarboxamide HEAs exhibited potent BACE-1 enzyme inhibition, but displayed poor cellular activity. Improvements in cellular activity and aspartic protease selectivity were observed for 7-azaindole-1,3-dicarboxamide HEAs. A methylprolinol-bearing derivative (10n) demonstrated robust reductions in rat plasma Aß levels, but did not lower rat brain Aß due to poor central exposure. The same analog exhibited a high efflux ratio in a bidirectional Caco-2 assay and was likely a substrate of the efflux transporter P-glycoprotein. X-ray crystal structures are reported for two indole HEAs in complex with BACE-1.


Subject(s)
Amines/chemistry , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Indoles/chemical synthesis , Protease Inhibitors/chemistry , Pyridines/chemical synthesis , Amines/chemical synthesis , Amines/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/blood , Animals , Aspartic Acid Endopeptidases/metabolism , Binding Sites , Crystallography, X-Ray , Indoles/chemistry , Indoles/pharmacology , Protease Inhibitors/chemical synthesis , Protease Inhibitors/pharmacology , Protein Structure, Tertiary , Pyridines/chemistry , Pyridines/pharmacology , Rats , Structure-Activity Relationship
16.
BMC Neurosci ; 11: 143, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-21054826

ABSTRACT

BACKGROUND: Accumulation of amyloid-ß (Aß) peptide in the brain is thought to play a key pathological role in Alzheimer's disease. Many pharmacological targets have therefore been proposed based upon the biochemistry of Aß, but not all are equally tractable for drug discovery. RESULTS: To search for novel targets that affect brain Aß without causing toxicity, we screened mouse brain samples from 1930 novel gene knock-out (KO) strains, representing 1926 genes, using Aß ELISA assays. Although robust Aß lowering was readily apparent in brains from a BACE1 KO strain, none of the novel strains exhibited robust decreases in brain Aß, including a GPR3 KO strain, which had previously been proposed as an Aß target. However, significantly increased Aß was observed in brain samples from two KO strains, corresponding to genes encoding the glycosylphosphatidylinositol mannosyl transferase PIGZ and quinolinate phosphoribosyltransferase (QPRT). CONCLUSIONS: Thus, gene ablations that are permissive for mouse survival and that also have a robust effect on Aß levels in the brain are rare.


Subject(s)
Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/genetics , Gene Knockout Techniques/methods , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/biosynthesis , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Genetic Testing/methods , Mannosyltransferases/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Pentosyltransferases/genetics , Peptide Fragments/biosynthesis , Peptide Fragments/metabolism
17.
ACS Med Chem Lett ; 1(3): 120-4, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-24900185

ABSTRACT

During the course of our research efforts to develop a potent and selective γ-secretase inhibitor for the treatment of Alzheimer's disease, we investigated a series of carboxamide-substituted sulfonamides. Optimization based on potency, Notch/amyloid-ß precursor protein selectivity, and brain efficacy after oral dosing led to the discovery of 4 (BMS-708163). Compound 4 is a potent inhibitor of γ-secretase (Aß40 IC50 = 0.30 nM), demonstrating a 193-fold selectivity against Notch. Oral administration of 4 significantly reduced Aß40 levels for sustained periods in brain, plasma, and cerebrospinal fluid in rats and dogs.

18.
J Biol Chem ; 283(34): 22992-3003, 2008 Aug 22.
Article in English | MEDLINE | ID: mdl-18574238

ABSTRACT

The amyloid-beta (Abeta) peptide, which likely plays a key role in Alzheimer disease, is derived from the amyloid-beta precursor protein (APP) through consecutive proteolytic cleavages by beta-site APP-cleaving enzyme and gamma-secretase. Unexpectedly gamma-secretase inhibitors can increase the secretion of Abeta peptides under some circumstances. This "Abeta rise" phenomenon, the same inhibitor causing an increase in Abeta at low concentrations but inhibition at higher concentrations, has been widely observed. Here we show that the Abeta rise depends on the beta-secretase-derived C-terminal fragment of APP (betaCTF) or C99 levels with low levels causing rises. In contrast, the N-terminally truncated form of Abeta, known as "p3," formed by alpha-secretase cleavage, did not exhibit a rise. In addition to the Abeta rise, low betaCTF or C99 expression decreased gamma-secretase inhibitor potency. This "potency shift" may be explained by the relatively high enzyme to substrate ratio under conditions of low substrate because increased concentrations of inhibitor would be necessary to affect substrate turnover. Consistent with this hypothesis, gamma-secretase inhibitor radioligand occupancy studies showed that a high level of occupancy was correlated with inhibition of Abeta under conditions of low substrate expression. The Abeta rise was also observed in rat brain after dosing with the gamma-secretase inhibitor BMS-299897. The Abeta rise and potency shift are therefore relevant factors in the development of gamma-secretase inhibitors and can be evaluated using appropriate choices of animal and cell culture models. Hypothetical mechanisms for the Abeta rise, including the "incomplete processing" and endocytic models, are discussed.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Gene Expression Regulation, Enzymologic , Animals , Brain/metabolism , Butyrates/pharmacology , Cell Line , Enzyme Inhibitors/pharmacology , Female , Humans , Hydrocarbons, Halogenated/pharmacology , Mice , Protein Binding , Protein Structure, Tertiary , Rats , Substrate Specificity
19.
J Pharmacol Exp Ther ; 326(2): 502-13, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18499745

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disease. Amyloid beta (Abeta) peptides are hypothesized to cause the initiation and progression of AD based on pathologic data from AD patients, genetic analysis of mutations that cause early onset forms of AD, and preclinical studies. Based on this hypothesis, beta-site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1) inhibitors are an attractive therapeutic approach for AD because cleavage of the APP by BACE1 is required to form Abeta. In this study, three potent BACE1 inhibitors are characterized. All three inhibitors decrease Abeta formation in cultured cells with IC(50) values less than 10 nM. Analysis of APP C-terminal fragments by immunoblotting and Abeta peptides by mass spectrometry showed that these inhibitors decreased Abeta by inhibiting BACE1. An assay for Abeta1-40 in mice was developed and used to show that these BACE1 inhibitors decreased plasma Abeta1-40, but not brain Abeta1-40, in wild-type mice. Because these BACE1 inhibitors were substrates for P-glycoprotein (P-gp), a member of the ATP-binding cassette superfamily of efflux transporters, these inhibitors were administered to P-gp knockout (KO) mice. These studies showed that all three BACE1 inhibitors decreased brain Abeta1-40 in P-gp KO mice, demonstrating that P-gp is a major limitation for development of BACE1 inhibitors to test the amyloid hypothesis. A comparison of plasma Abeta1-40 and brain Abeta1-40 dose responses for these three compounds revealed differences in relative ED(50) values, indicating that factors other than P-gp can also contribute to poor brain activity by BACE1 inhibitors.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/antagonists & inhibitors , Brain , Enzyme Inhibitors/pharmacology , Peptide Fragments/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Peptides/blood , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases/physiology , Blotting, Western , Brain/drug effects , Brain/enzymology , Brain/metabolism , Cell Line , Cell Membrane Permeability , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Mice, Knockout , Molecular Structure , Peptide Fragments/blood , Protein Binding , Substrate Specificity
20.
J Neurosci Methods ; 168(2): 465-74, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18187205

ABSTRACT

Targeting the metabolism of amyloid beta peptides (Abeta) is currently the leading experimental approach to treatment of Alzheimer's disease (AD). Described here is an immunoprecipitation-liquid chromatography/mass spectrometry (ip-LC/MS) assay to simultaneously characterize and quantitate different forms of Abeta in biological samples. The 4G8 antibody, specific for the 17-24 amino acid epitope of Abeta was employed to selectively isolate Abeta from in vitro samples for subsequent LC-MS analysis. A high resolution accurate mass hybrid linear ion trap-Orbitrap, LTQ-Orbitrap mass spectrometer was used to identify forms of 12 Abeta in H4-APP751 swe cell extracts based on ab initio calculations, accurate mass measurements, isotopic modeling and by de novo peptide sequencing using tandem mass spectrometry. The quantitative LC-MS analysis was performed on a linear ion trap mass spectrometer, LTQ, in full scan mode, this mode of operation enables sensitive detection levels and post-acquisition data mining for different forms of Abeta for quantitative assessment. Dosing studies with three known inhibitors of Abeta production, sulindac sulfide (SSide), BMS-299897 ('897) and compound W (CW) are reported to demonstrate the utility and analytical characteristics of the assay. This assay has the potential to provide insight into the formation of Abeta; increase understanding of drug mechanisms; and to contribute to drug efficacy studies.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amino Acid Sequence , Amyloid beta-Peptides/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Blotting, Western , Butyrates/pharmacology , Cell Line , Chromatography, High Pressure Liquid , Data Interpretation, Statistical , Diiodothyronines/pharmacology , Enzyme-Linked Immunosorbent Assay , Humans , Hydrocarbons, Halogenated/pharmacology , Immunoprecipitation , Linear Models , Mass Spectrometry , Molecular Sequence Data , Reference Standards , Reproducibility of Results , Sulindac/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...