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










Publication year range
1.
ACS Med Chem Lett ; 12(9): 1389-1395, 2021 Sep 09.
Article in English | MEDLINE | ID: mdl-34531947

ABSTRACT

An unbiased phenotypic neuronal assay was developed to measure the synaptotoxic effects of soluble Aß oligomers. A collection of CNS druglike small molecules prepared by conditioned extraction was screened. Compounds that prevented and reversed synaptotoxic effects of Aß oligomers in neurons were discovered to bind to the sigma-2 receptor complex. Select development compounds displaced receptor-bound Aß oligomers, rescued synapses, and restored cognitive function in transgenic hAPP Swe/Ldn mice. Our first-in-class orally administered small molecule investigational drug 7 (CT1812) has been advanced to Phase II clinical studies for Alzheimer's disease.

2.
Alzheimers Dement ; 17(8): 1365-1382, 2021 08.
Article in English | MEDLINE | ID: mdl-33559354

ABSTRACT

INTRODUCTION: Amyloid beta (Aß) oligomers are one of the most toxic structural forms of the Aß protein and are hypothesized to cause synaptotoxicity and memory failure as they build up in Alzheimer's disease (AD) patients' brain tissue. We previously demonstrated that antagonists of the sigma-2 receptor complex effectively block Aß oligomer toxicity. CT1812 is an orally bioavailable, brain penetrant small molecule antagonist of the sigma-2 receptor complex that appears safe and well tolerated in healthy elderly volunteers. We tested CT1812's effect on Aß oligomer pathobiology in preclinical AD models and evaluated CT1812's impact on cerebrospinal fluid (CSF) protein biomarkers in mild to moderate AD patients in a clinical trial (ClinicalTrials.gov NCT02907567). METHODS: Experiments were performed to measure the impact of CT1812 versus vehicle on Aß oligomer binding to synapses in vitro, to human AD patient post mortem brain tissue ex vivo, and in living APPSwe /PS1dE9 transgenic mice in vivo. Additional experiments were performed to measure the impact of CT1812 versus vehicle on Aß oligomer-induced deficits in membrane trafficking rate, synapse number, and protein expression in mature hippocampal/cortical neurons in vitro. The impact of CT1812 on cognitive function was measured in transgenic Thy1 huAPPSwe/Lnd+ and wild-type littermates. A multicenter, double-blind, placebo-controlled parallel group trial was performed to evaluate the safety, tolerability, and impact on protein biomarker expression of CT1812 or placebo given once daily for 28 days to AD patients (Mini-Mental State Examination 18-26). CSF protein expression was measured by liquid chromatography with tandem mass spectrometry or enzyme-linked immunosorbent assay in samples drawn prior to dosing (Day 0) and at end of dosing (Day 28) and compared within each patient and between pooled treated versus placebo-treated dosing groups. RESULTS: CT1812 significantly and dose-dependently displaced Aß oligomers bound to synaptic receptors in three independent preclinical models of AD, facilitated oligomer clearance into the CSF, increased synaptic number and protein expression in neurons, and improved cognitive performance in transgenic mice. CT1812 significantly increased CSF concentrations of Aß oligomers in AD patient CSF, reduced concentrations of synaptic proteins and phosphorylated tau fragments, and reversed expression of many AD-related proteins dysregulated in CSF. DISCUSSION: These preclinical studies demonstrate the novel disease-modifying mechanism of action of CT1812 against AD and Aß oligomers. The clinical results are consistent with preclinical data and provide evidence of target engagement and impact on fundamental disease-related signaling pathways in AD patients, supporting further development of CT1812.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Cognition/drug effects , Mice, Transgenic , Receptors, sigma/antagonists & inhibitors , Aged , Animals , Brain/metabolism , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Hippocampus/metabolism , Humans , Male , Mice , Neurons/metabolism , Synapses/metabolism
3.
J Neurosci Res ; 99(4): 1161-1176, 2021 04.
Article in English | MEDLINE | ID: mdl-33480104

ABSTRACT

α-Synuclein oligomers are thought to have a pivotal role in sporadic and familial Parkinson's disease (PD) and related α-synucleinopathies, causing dysregulation of protein trafficking, autophagy/lysosomal function, and protein clearance, as well as synaptic function impairment underlying motor and cognitive symptoms of PD. Moreover, trans-synaptic spread of α-synuclein oligomers is hypothesized to mediate disease progression. Therapeutic approaches that effectively block α-synuclein oligomer-induced pathogenesis are urgently needed. Here, we show for the first time that α-synuclein species isolated from human PD patient brain and recombinant α-synuclein oligomers caused similar deficits in lipid vesicle trafficking rates in cultured rat neurons and glia, while α-synuclein species isolated from non-PD human control brain samples did not. Recombinant α-synuclein oligomers also increased neuronal expression of lysosomal-associated membrane protein-2A (LAMP-2A), the lysosomal receptor that has a critical role in chaperone-mediated autophagy. Unbiased screening of several small molecule libraries (including the NIH Clinical Collection) identified sigma-2 receptor antagonists as the most effective at blocking α-synuclein oligomer-induced trafficking deficits and LAMP-2A upregulation in a dose-dependent manner. These results indicate that antagonists of the sigma-2 receptor complex may alleviate α-synuclein oligomer-induced neurotoxicity and are a novel therapeutic approach for disease modification in PD and related α-synucleinopathies.


Subject(s)
Parkinson Disease/metabolism , Receptors, sigma/antagonists & inhibitors , Receptors, sigma/metabolism , alpha-Synuclein/metabolism , Aged , Aged, 80 and over , Animals , Autophagy/drug effects , Brain/metabolism , Female , High-Throughput Screening Assays , Humans , Lipid Metabolism , Lysosomal-Associated Membrane Protein 2/metabolism , Male , Parkinson Disease/pathology , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Vesicular Transport Proteins/metabolism , alpha-Synuclein/pharmacology
4.
PLoS One ; 9(11): e111898, 2014.
Article in English | MEDLINE | ID: mdl-25390368

ABSTRACT

Synaptic dysfunction and loss caused by age-dependent accumulation of synaptotoxic beta amyloid (Abeta) 1-42 oligomers is proposed to underlie cognitive decline in Alzheimer's disease (AD). Alterations in membrane trafficking induced by Abeta oligomers mediates reduction in neuronal surface receptor expression that is the basis for inhibition of electrophysiological measures of synaptic plasticity and thus learning and memory. We have utilized phenotypic screens in mature, in vitro cultures of rat brain cells to identify small molecules which block or prevent the binding and effects of Abeta oligomers. Synthetic Abeta oligomers bind saturably to a single site on neuronal synapses and induce deficits in membrane trafficking in neuronal cultures with an EC50 that corresponds to its binding affinity. The therapeutic lead compounds we have found are pharmacological antagonists of Abeta oligomers, reducing the binding of Abeta oligomers to neurons in vitro, preventing spine loss in neurons and preventing and treating oligomer-induced deficits in membrane trafficking. These molecules are highly brain penetrant and prevent and restore cognitive deficits in mouse models of Alzheimer's disease. Counter-screening these compounds against a broad panel of potential CNS targets revealed they are highly potent and specific ligands of the sigma-2/PGRMC1 receptor. Brain concentrations of the compounds corresponding to greater than 80% receptor occupancy at the sigma-2/PGRMC1 receptor restore cognitive function in transgenic hAPP Swe/Ldn mice. These studies demonstrate that synthetic and human-derived Abeta oligomers act as pharmacologically-behaved ligands at neuronal receptors--i.e. they exhibit saturable binding to a target, they exert a functional effect related to their binding and their displacement by small molecule antagonists blocks their functional effect. The first-in-class small molecule receptor antagonists described here restore memory to normal in multiple AD models and sustain improvement long-term, representing a novel mechanism of action for disease-modifying Alzheimer's therapeutics.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/chemistry , Neurons/metabolism , Peptide Fragments/chemistry , Synapses/drug effects , Alzheimer Disease/metabolism , Animals , Brain/drug effects , Chemistry, Pharmaceutical , Cognition/drug effects , Cognition Disorders/drug therapy , Drug Design , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Mice, Transgenic , Neuroglia/metabolism , Protein Binding , Protein Transport , Rats , Rats, Sprague-Dawley , Synapses/metabolism
5.
PLoS One ; 9(11): e111899, 2014.
Article in English | MEDLINE | ID: mdl-25390692

ABSTRACT

Amyloid beta (Abeta) 1-42 oligomers accumulate in brains of patients with Mild Cognitive Impairment (MCI) and disrupt synaptic plasticity processes that underlie memory formation. Synaptic binding of Abeta oligomers to several putative receptor proteins is reported to inhibit long-term potentiation, affect membrane trafficking and induce reversible spine loss in neurons, leading to impaired cognitive performance and ultimately to anterograde amnesia in the early stages of Alzheimer's disease (AD). We have identified a receptor not previously associated with AD that mediates the binding of Abeta oligomers to neurons, and describe novel therapeutic antagonists of this receptor capable of blocking Abeta toxic effects on synapses in vitro and cognitive deficits in vivo. Knockdown of sigma-2/PGRMC1 (progesterone receptor membrane component 1) protein expression in vitro using siRNA results in a highly correlated reduction in binding of exogenous Abeta oligomers to neurons of more than 90%. Expression of sigma-2/PGRMC1 is upregulated in vitro by treatment with Abeta oligomers, and is dysregulated in Alzheimer's disease patients' brain compared to age-matched, normal individuals. Specific, high affinity small molecule receptor antagonists and antibodies raised against specific regions on this receptor can displace synthetic Abeta oligomer binding to synaptic puncta in vitro and displace endogenous human AD patient oligomers from brain tissue sections in a dose-dependent manner. These receptor antagonists prevent and reverse the effects of Abeta oligomers on membrane trafficking and synapse loss in vitro and cognitive deficits in AD mouse models. These findings suggest sigma-2/PGRMC1 receptors mediate saturable oligomer binding to synaptic puncta on neurons and that brain penetrant, small molecules can displace endogenous and synthetic oligomers and improve cognitive deficits in AD models. We propose that sigma-2/PGRMC1 is a key mediator of the pathological effects of Abeta oligomers in AD and is a tractable target for small molecule disease-modifying therapeutics.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/chemistry , Membrane Proteins/metabolism , Peptide Fragments/chemistry , Receptors, Progesterone/metabolism , Synapses/drug effects , Alzheimer Disease/metabolism , Animals , Autoradiography , Brain/metabolism , Cell Membrane/metabolism , Cognition/drug effects , Cognition Disorders/drug therapy , Humans , Membrane Proteins/genetics , Mice , Neurons/metabolism , Protein Binding , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Progesterone/genetics , Synapses/metabolism
6.
Sci China Life Sci ; 53(1): 94-100, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20596960

ABSTRACT

High mammalian gene expression was obtained for more than twenty different proteins in different cell types by just a few laboratory scale stable gene transfections for each protein. The stable expression vectors were constructed by inserting a naturally-occurring 1.006 kb or a synthetic 0.733 kb DNA fragment (including intron) of extremely GC-rich at the 5' or/and 3' flanking regions of these protein genes or their gene promoters. This experiment is the first experimental evidence showing that a non-coding extremely GC-rich DNA fragment is a super "chromatin opening element" and plays an important role in mammalian gene expression. This experiment has further indicated that chromatin-based regulation of mammalian gene expression is at least partially embedded in DNA primary structure, namely DNA GC-content.


Subject(s)
Base Composition/genetics , DNA/genetics , Gene Expression , Introns/genetics , Actins/genetics , Animals , Base Sequence , CHO Cells , Cells, Cultured , Chickens , Chromatin/genetics , Cricetinae , Cricetulus , Mammals/genetics , Molecular Sequence Data , Transfection
7.
Nat Chem Biol ; 5(7): 441-7, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19536101

ABSTRACT

Between 2004 and 2008, the US National Institutes of Health Molecular Libraries and Imaging initiative pilot phase funded 10 high-throughput screening centers, resulting in the deposition of 691 assays into PubChem and the nomination of 64 chemical probes. We crowdsourced the Molecular Libraries and Imaging initiative output to 11 experts, who expressed medium or high levels of confidence in 48 of these 64 probes.


Subject(s)
Drug Discovery/methods , Molecular Probe Techniques/trends , Molecular Probes/chemistry , Small Molecule Libraries/chemistry , Databases, Factual , Decision Making , Drug Discovery/economics , Drug Discovery/organization & administration , Drug Discovery/standards , Molecular Probe Techniques/standards , National Institutes of Health (U.S.) , United States
8.
Recent Pat CNS Drug Discov ; 3(3): 200-8, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18991809

ABSTRACT

Small molecule modulators of neural stem cell (NSC) differentiation might potentially be developed into orally administered neurogenic drugs to treat neurodegenerative diseases including Alzheimer's disease. New technologies developed for the study of NSC culture, proliferation and differentiation have enabled the establishment of screening platforms to identify small molecules with neurogenic activity. Recent patents claim novel small molecules identified from screening collections that stimulate or otherwise regulate stem cell differentiation and neurogenesis. Several patents claim newly discovered NSC differentiation modulating activity of previously marketed drugs suggesting perhaps a previously unknown mechanism of action of these drugs and/or implicating the target enzyme and receptor pathways as key players in neurogenesis. This relatively new area of research into small molecule modulators of neurogenesis is reviewed and recent patents claiming small molecule neurogenic compounds, potentially orally administered CNS regenerative therapies are summarized.


Subject(s)
Neurogenesis/drug effects , Animals , Cells, Cultured , Drug Evaluation, Preclinical , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neurons/drug effects , Neurons/physiology , PPAR gamma/drug effects , Receptors, Serotonin/drug effects , Stem Cells/cytology , Stem Cells/drug effects
9.
Am J Cardiol ; 101(10A): 43D-49D, 2008 May 22.
Article in English | MEDLINE | ID: mdl-18474274

ABSTRACT

The history of drug development has its foundation firmly set in the study of natural remedies used to treat human disease over centuries. Analysis of medicinal plants, bioactive cultures, and increased understanding of micronutrients in the food chain opened the door to the development of purified and defined chemical compounds as dose-controlled medicines. Thus, with the early discovery of cardiotonics in foxglove, salicylic acid in willow bark, morphine in poppies, and penicillin in mold, the pharmaceutical industry was launched. Such natural small molecules served as treatments for disease and ultimately, as pharmacologic tools to enable the understanding of the biochemical pathways and mechanisms of disease. In contrast, modern drug discovery technologies coupled with the powerful tools of biotechnology have prompted drug discovery organizations to focus on target-driven drug discovery at the molecular level by launching high-throughput screening programs using artificial biochemical assays. At a time when the pharmaceutical industry has come under scrutiny for high rates of drug development failure, it is interesting to see that natural products drug discovery has been marginalized in favor of this high-throughput biochemical screening paradigm. If modern drug development is once again to benefit from natural products as a source, then the limitations of artificial biochemical assays as applied to the screening of natural extracts must be realized in order to capitalize on the vast natural molecular diversity and rich ethnobotanic data that has emerged worldwide. Natural compounds can again become central players in the treatment of disease and in the understanding of disease mechanisms.


Subject(s)
Drug Design , Plant Extracts/chemistry , Plants, Medicinal/chemistry , Biological Products , Drug Industry/history , Ethnobotany/trends , History, 18th Century , History, 19th Century , History, 20th Century , Humans , Plant Extracts/history
10.
Curr Opin Chem Biol ; 12(3): 340-51, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18328272

ABSTRACT

Molecular diversity is of vital importance in drug screening in general and for the discovery and development of new pharmacophores in particular. Biochemical screening is a powerful tool for pharmacophore development given understanding of the properties of a good lead compound operating in the biochemical environment. The properties of leadlikeness have evolved to accommodate the artificial conditions of a biochemical assay. Accordingly, the properties of leadlikeness that are suited for screening at protein targets biochemically are different and complementary to the properties of druglikeness used to guide the selection of good compounds studied biologically in cellular studies and animal models. The benefits of leadlikeness in the biochemical screening arena (including fragment-based screening and co-crystallization studies) are described here and recommendations are forwarded for the generation of leadlike molecular diversity. Chemically stable low molecular weight 'minimalist' compounds (or fragments) with dense heteroatom substitution and variable conformational constraint are promoted as conceptually superior compounds for biochemical screening.


Subject(s)
Drug Evaluation, Preclinical/methods , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism , Biological Products/chemistry , Biological Products/metabolism , Biological Products/pharmacology , Molecular Conformation , Nitrogen/chemistry , Proteins/metabolism
12.
Curr Opin Drug Discov Devel ; 9(3): 303-13, 2006 May.
Article in English | MEDLINE | ID: mdl-16729726

ABSTRACT

This review summarizes progress made in the development of fully computational approaches to the prediction of blood-brain barrier (BBB) permeability of small molecules, with a focus on rapid computational methods suitable for the analysis of large compound sets and virtual screening. A comparative analysis using the recently developed Advanced Chemistry Development (ACD/Labs) Inc BBB permeability algorithm for the calculation of logBB values for known Alzheimer's disease medicines, selected central nervous system drugs and new secretase inhibitors for Alzheimer's disease, is presented. The trends in logBB values and the associated physiochemical properties of these agents as they relate to the potential for BBB permeability are also discussed.


Subject(s)
Alzheimer Disease/drug therapy , Blood-Brain Barrier/physiology , Central Nervous System Agents/metabolism , Computer Simulation , Endopeptidases/metabolism , Protease Inhibitors/metabolism , Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , Biological Transport , Brain/metabolism , Capillary Permeability , Humans
13.
Med Chem ; 1(5): 519-27, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16787336

ABSTRACT

The pharmaceutical industry currently suffers unsustainably high program failure rates despite our best efforts to implement drug design methods and to develop high throughput biochemical screening technologies over the past 20 years. While much of this failure is rationalized to be due to uncontrollable late stage drug development issues and clinical events, it has become increasingly clear that the choices we make in early drug discovery are vital to the ultimate failure or success outcomes of our drug discovery programs. The judicious selection of high probability of success therapeutic modalities, the rigorous determination of leadlikeness and druglikeness, and the all-important selection of high probability of success enzyme and receptor targets are the vital drivers of failure and success in small molecule drug discovery as it is performed in the age of biochemical screening. Consideration of these guiding principles will improve our chances of success in drug discovery, and increase our ability to address unmet medical need in the future.


Subject(s)
Drug Design , Drug Evaluation, Preclinical/methods , Drug Industry/methods , Drug Industry/organization & administration , Clinical Trials as Topic , Decision Making, Organizational , Research Design
14.
J Pharmacol Exp Ther ; 308(2): 627-35, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14593085

ABSTRACT

Calcimimetic compounds, which activate the parathyroid cell Ca(2+) receptor (CaR) and inhibit parathyroid hormone (PTH) secretion, are under experimental study as a treatment for hyperparathyroidism. This report describes the salient pharmacodynamic properties, using several test systems, of a new calcimimetic compound, cinacalcet HCl. Cinacalcet HCl increased the concentration of cytoplasmic Ca(2+) ([Ca(2+)](i)) in human embryonic kidney 293 cells expressing the human parathyroid CaR. Cinacalcet HCl (EC(50) = 51 nM) in the presence of 0.5 mM extracellular Ca(2+) elicited increases in [Ca(2+)](i) in a dose- and calcium-dependent manner. Similarly, in the presence of 0.5 mM extracellular Ca(2+), cinacalcet HCl (IC(50) = 28 nM) produced a concentration-dependent decrease in PTH secretion from cultured bovine parathyroid cells. Using rat medullary thyroid carcinoma 6-23 cells expressing the CaR, cinacalcet HCl (EC(50) = 34 nM) produced a concentration-dependent increase in calcitonin secretion. In vivo studies in rats demonstrated cinacalcet HCl is orally bioavailable and displays approximately linear pharmacokinetics over the dose range of 1 to 36 mg/kg. Furthermore, this compound suppressed serum PTH and blood-ionized Ca(2+) levels and increased serum calcitonin levels in a dose-dependent manner. Cinacalcet was about 30-fold more potent at lowering serum levels of PTH than it was at increasing serum calcitonin levels. The S-enantiomer of cinacalcet (S-AMG 073) was at least 75-fold less active in these assay systems. The present findings provide compelling evidence that cinacalcet HCl is a potent and stereoselective activator of the parathyroid CaR and, as such, might be beneficial in the treatment of hyperparathyroidism.


Subject(s)
Calcitonin/metabolism , Naphthalenes/pharmacology , Parathyroid Glands/drug effects , Parathyroid Hormone/metabolism , Animals , Calcitonin/blood , Calcium/blood , Calcium-Binding Proteins/metabolism , Cells, Cultured , Cinacalcet , Humans , Male , Naphthalenes/pharmacokinetics , Parathyroid Glands/metabolism , Parathyroid Hormone/blood , Phosphorus/blood , Rats , Rats, Sprague-Dawley
15.
Drug Discov Today ; 8(2): 86-96, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12565011

ABSTRACT

Biochemical assays have largely supplanted functional biological assays as drug screening tools in the early stages of drug discovery. The de-selection of compounds that are 'nonleadlike' binders (and bonders) and the proactive selection of those compounds that are 'leadlike' in their binding to the target are vital components of the screening effort. The physiochemical properties of leadlikeness and the surprising differences between those properties and the now classical definitions of druglikeness are becoming apparent.


Subject(s)
Chemistry, Pharmaceutical , Drug Evaluation, Preclinical/methods , Structure-Activity Relationship , Chemistry, Pharmaceutical/methods , Chemistry, Pharmaceutical/organization & administration , Chemistry, Pharmaceutical/trends
SELECTION OF CITATIONS
SEARCH DETAIL
...