Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
2.
Nature ; 537(7618): 50-6, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27582220

ABSTRACT

Alzheimer's disease (AD) is characterized by deposition of amyloid-ß (Aß) plaques and neurofibrillary tangles in the brain, accompanied by synaptic dysfunction and neurodegeneration. Antibody-based immunotherapy against Aß to trigger its clearance or mitigate its neurotoxicity has so far been unsuccessful. Here we report the generation of aducanumab, a human monoclonal antibody that selectively targets aggregated Aß. In a transgenic mouse model of AD, aducanumab is shown to enter the brain, bind parenchymal Aß, and reduce soluble and insoluble Aß in a dose-dependent manner. In patients with prodromal or mild AD, one year of monthly intravenous infusions of aducanumab reduces brain Aß in a dose- and time-dependent manner. This is accompanied by a slowing of clinical decline measured by Clinical Dementia Rating-Sum of Boxes and Mini Mental State Examination scores. The main safety and tolerability findings are amyloid-related imaging abnormalities. These results justify further development of aducanumab for the treatment of AD. Should the slowing of clinical decline be confirmed in ongoing phase 3 clinical trials, it would provide compelling support for the amyloid hypothesis.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/psychology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal, Humanized/therapeutic use , Plaque, Amyloid/drug therapy , Plaque, Amyloid/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid/drug effects , Amyloid/metabolism , Amyloid beta-Peptides/chemistry , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Brain/drug effects , Brain/metabolism , Clinical Trials, Phase III as Topic , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Models, Biological , Plaque, Amyloid/pathology , Protein Aggregation, Pathological/drug therapy , Solubility
3.
Neuropharmacology ; 103: 57-68, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26690893

ABSTRACT

Reducing the production of larger aggregation-prone amyloid ß-peptides (Aß) remains an untested therapeutic approach for reducing the appearance and growth of Aß plaques in the brain, which are a hallmark pathological feature of Alzheimer's disease. γ-Secretase modulators (GSMs) are therapeutics that impact γ-secretase-dependent cleavage of amyloid precursor protein to promote the production of shorter Aß peptides that are less prone to aggregation and plaque deposition. This is accomplished without inhibiting overall γ-secretase function and cleavage of other substrates, which is believed to be a source of deleterious side effects. Here, we report the pharmacokinetic and pharmacodynamic properties of BIIB042, a novel bioavailable and brain-penetrant GSM. In cell-based assays, BIIB042 reduced the levels of Aß42, increased the levels of Aß38 and had little effect on the levels of Aß40, the most abundant Aß species. Similar pharmacodynamic properties were confirmed in the central nervous system and in plasma of mice and rats, and also in plasma of cynomolgus monkeys after a single oral dose of BIIB042. BIIB042 reduced Aß42 levels and Aß plaque burden in Tg2576 mice, which overexpress human amyloid precursor protein and serve as a model system for Alzheimer's disease. BIIB042 did not inhibit cleavage of other γ-secretase substrates in cell-based and in vivo signaling and cleavage assays. The pharmacodynamic effects of lowering Aß42 in the central nervous system coupled with demonstrated efficacy in reducing plaque pathology suggests modulation of γ-secretase, with molecules like BIIB042, is a compelling therapeutic approach for the treatment of Alzheimer's disease.


Subject(s)
Aldehydes/pharmacokinetics , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Brain/drug effects , Brain/enzymology , Aldehydes/administration & dosage , Amyloid beta-Peptides/blood , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Macaca fascicularis , Male , Mice , Plaque, Amyloid/metabolism , Protein Isoforms/blood , Rats , Rats, Inbred F344
4.
J Pharmacol Exp Ther ; 341(1): 274-84, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22267202

ABSTRACT

Oxidative stress is central to the pathology of several neurodegenerative diseases, including multiple sclerosis, and therapeutics designed to enhance antioxidant potential could have clinical value. The objective of this study was to characterize the potential direct neuroprotective effects of dimethyl fumarate (DMF) and its primary metabolite monomethyl fumarate (MMF) on cellular resistance to oxidative damage in primary cultures of central nervous system (CNS) cells and further explore the dependence and function of the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) pathway in this process. Treatment of animals or primary cultures of CNS cells with DMF or MMF resulted in increased nuclear levels of active Nrf2, with subsequent up-regulation of canonical antioxidant target genes. DMF-dependent up-regulation of antioxidant genes in vivo was lost in mice lacking Nrf2 [Nrf2(-/-)]. DMF or MMF treatment increased cellular redox potential, glutathione, ATP levels, and mitochondrial membrane potential in a concentration-dependent manner. Treating astrocytes or neurons with DMF or MMF also significantly improved cell viability after toxic oxidative challenge in a concentration-dependent manner. This effect on viability was lost in cells that had eliminated or reduced Nrf2. These data suggest that DMF and MMF are cytoprotective for neurons and astrocytes against oxidative stress-induced cellular injury and loss, potentially via up-regulation of an Nrf2-dependent antioxidant response. These data also suggest DMF and MMF may function through improving mitochondrial function. The clinical utility of DMF in multiple sclerosis is being explored through phase III trials with BG-12, which is an oral therapeutic containing DMF as the active ingredient.


Subject(s)
Central Nervous System/cytology , Central Nervous System/metabolism , Cytoprotection/genetics , Fumarates/pharmacology , NF-E2-Related Factor 2/physiology , Neurons/metabolism , Oxidative Stress/genetics , Signal Transduction/genetics , Animals , Cells, Cultured , Central Nervous System/drug effects , Cytoprotection/drug effects , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/deficiency , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Signal Transduction/drug effects
5.
Bioorg Med Chem Lett ; 21(24): 7277-80, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22061640

ABSTRACT

Starting from literature examples of nonsteroidal anti-inflammatory drugs (NSAIDs)-type carboxylic acid γ-secretase modulators (GSMs) and using a scaffold design approach, we identified 4-aminomethylphenylacetic acid 4 with a desirable γ-secretase modulation profile. Scaffold optimization led to the discovery of a novel chemical series, represented by 6b, having improved brain penetration. Further SAR studies provided analog 6q that exhibited a good pharmacological profile. Oral administration of 6q significantly reduced brain Aß42 levels in mice and rats.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Enzyme Inhibitors/chemistry , Phenylacetates/chemistry , Piperidines/chemistry , Administration, Oral , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Brain/metabolism , Drug Design , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Mice , Peptide Fragments/metabolism , Phenylacetates/chemical synthesis , Phenylacetates/pharmacokinetics , Piperidines/chemical synthesis , Piperidines/pharmacokinetics , Rats
6.
Brain ; 134(Pt 3): 678-92, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21354971

ABSTRACT

Inflammation and oxidative stress are thought to promote tissue damage in multiple sclerosis. Thus, novel therapeutics enhancing cellular resistance to free radicals could prove useful for multiple sclerosis treatment. BG00012 is an oral formulation of dimethylfumarate. In a phase II multiple sclerosis trial, BG00012 demonstrated beneficial effects on relapse rate and magnetic resonance imaging markers indicative of inflammation as well as axonal destruction. First we have studied effects of dimethylfumarate on the disease course, central nervous system, tissue integrity and the molecular mechanism of action in an animal model of chronic multiple sclerosis: myelin oligodendrocyte glycoprotein induced experimental autoimmune encephalomyelitis in C57BL/6 mice. In the chronic phase of experimental autoimmune encephalomyelitis, preventive or therapeutic application of dimethylfumarate ameliorated the disease course and improved preservation of myelin, axons and neurons. In vitro, the application of fumarates increased murine neuronal survival and protected human or rodent astrocytes against oxidative stress. Application of dimethylfumarate led to stabilization of the transcription factor nuclear factor (erythroid-derived 2)-related factor 2, activation of nuclear factor (erythroid-derived 2)-related factor 2-dependent transcriptional activity and accumulation of NADP(H) quinoline oxidoreductase-1 as a prototypical target gene. Furthermore, the immediate metabolite of dimethylfumarate, monomethylfumarate, leads to direct modification of the inhibitor of nuclear factor (erythroid-derived 2)-related factor 2, Kelch-like ECH-associated protein 1, at cysteine residue 151. In turn, increased levels of nuclear factor (erythroid-derived 2)-related factor 2 and reduced protein nitrosylation were detected in the central nervous sytem of dimethylfumarate-treated mice. Nuclear factor (erythroid-derived 2)-related factor 2 was also upregulated in the spinal cord of autopsy specimens from untreated patients with multiple sclerosis. In dimethylfumarate-treated mice suffering from experimental autoimmune encephalomyelitis, increased immunoreactivity for nuclear factor (erythroid-derived 2)-related factor 2 was detected by confocal microscopy in neurons of the motor cortex and the brainstem as well as in oligodendrocytes and astrocytes. In mice deficient for nuclear factor (erythroid-derived 2)-related factor 2 on the same genetic background, the dimethylfumarate mediated beneficial effects on clinical course, axon preservation and astrocyte activation were almost completely abolished thus proving the functional relevance of this transcription factor for the neuroprotective mechanism of action. We conclude that the ability of dimethylfumarate to activate nuclear factor (erythroid-derived 2)-related factor 2 may offer a novel cytoprotective modality that further augments the natural antioxidant responses in multiple sclerosis tissue and is not yet targeted by other multiple sclerosis therapies.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/prevention & control , Fumarates/therapeutic use , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/therapeutic use , Signal Transduction/drug effects , 2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism , Aldehyde Reductase/metabolism , Animals , Antigens, Differentiation/metabolism , Antioxidants/metabolism , Astrocytes/drug effects , Astrocytes/immunology , Axons/metabolism , Axons/pathology , CD3 Complex/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid/methods , Cytokines/metabolism , Disease Models, Animal , Embryo, Mammalian , Encephalomyelitis, Autoimmune, Experimental/etiology , Female , Fumarates/pharmacology , Gene Expression Regulation/drug effects , Glycoproteins/adverse effects , Green Fluorescent Proteins/genetics , Humans , Hydrogen Peroxide/pharmacology , Mass Spectrometry , Mice , Mice, Inbred C57BL , Motor Neurons/cytology , Motor Neurons/drug effects , Motor Neurons/metabolism , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Myelin Proteins/metabolism , Myelin-Oligodendrocyte Glycoprotein , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neuroprotective Agents/pharmacology , Nogo Proteins , Oxidative Stress/drug effects , Peptide Fragments/adverse effects , RNA, Small Interfering/pharmacology , Sleep/physiology , Spinal Cord/cytology , Statistics, Nonparametric , Tandem Mass Spectrometry/methods , Time Factors , Transfection
7.
ACS Med Chem Lett ; 2(10): 786-91, 2011 Oct 13.
Article in English | MEDLINE | ID: mdl-24900267

ABSTRACT

We have investigated a novel series of acid-derived γ-secretase modulators as a potential treatment of Alzheimer's disease. Optimization based on cellular potency and brain pharmacodynamics after oral dosing led to the discovery of 10a (BIIB042). Compound 10a is a potent γ-secretase modulator, which lowered Aß42, increased Aß38, but had little to no effect on Aß40 levels both in vitro and in vivo. In addition, compound 10a did not affect Notch signaling in our in vitro assessment. Compound 10a demonstrated excellent pharmacokinetic parameters in multiple species. Oral administration of 10a significantly reduced brain Aß42 levels in CF-1 mice and Fischer rats, as well as plasma Aß42 levels in cynomolgus monkeys. Compound 10a was selected as a candidate for preclinical safety evaluation.

8.
Bioorg Med Chem Lett ; 20(7): 2264-9, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20188554

ABSTRACT

Modifying FTY720, an immunosuppressant modulator, led to a new series of well phosphorylated tetralin analogs as potent S1P1 receptor agonists. The stereochemistry effect of tetralin ring was probed, and (-)-(R)-2-amino-2-((S)-6-octyl-1,2,3,4-tetrahydronaphthalen-2-yl)propan-1-ol was identified as a good SphK2 substrate and potent S1P1 agonist with good oral bioavailability.


Subject(s)
Immunosuppressive Agents/pharmacology , Prodrugs/pharmacology , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/metabolism , Tetrahydronaphthalenes/pharmacology , Administration, Oral , Animals , Crystallography, X-Ray , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/metabolism , Immunosuppressive Agents/pharmacokinetics , Lymphopenia/chemically induced , Mice , Models, Molecular , Multiple Sclerosis/drug therapy , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Prodrugs/chemistry , Prodrugs/metabolism , Prodrugs/pharmacokinetics , Structure-Activity Relationship , Tetrahydronaphthalenes/chemistry , Tetrahydronaphthalenes/metabolism , Tetrahydronaphthalenes/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...