Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters











Database
Language
Publication year range
1.
ACS Chem Neurosci ; 10(3): 1765-1782, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30525452

ABSTRACT

In order to determine the contributions of histone deacetylase (HDAC) isoforms to the beneficial effects of dual phosphodiesterase 5 (PDE5) and pan-HDAC inhibitors on in vivo models of Alzheimer's disease (AD), we have designed, synthesized, and tested novel chemical probes with the desired target compound profile of PDE5 and class I HDAC selective inhibitors. Compared to previous hydroxamate-based series, these molecules exhibit longer residence times on HDACs. In this scenario, shorter or longer preincubation times may have a significant impact on the IC50 values of these compounds and therefore on their corresponding selectivity profiles on the different HDAC isoforms. On the other hand, different chemical series have been explored and, as expected, some pairwise comparisons show a clear impact of the scaffold on biological responses (e.g., 35a vs 40a). The lead identification process led to compound 29a, which shows an adequate ADME-Tox profile and in vivo target engagement (histone acetylation and cAMP/cGMP response element-binding (CREB) phosphorylation) in the central nervous system (CNS), suggesting that this compound represents an optimized chemical probe; thus, 29a has been assayed in a mouse model of AD (Tg2576).


Subject(s)
Alzheimer Disease/drug therapy , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Histone Deacetylase Inhibitors/pharmacology , Phosphodiesterase 5 Inhibitors/pharmacology , Acetylation/drug effects , Animals , Disease Models, Animal , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/drug effects , Histone Deacetylases/metabolism , Humans , Mice , Phosphodiesterase 5 Inhibitors/chemistry
2.
Eur J Med Chem ; 150: 506-524, 2018 Apr 25.
Article in English | MEDLINE | ID: mdl-29549837

ABSTRACT

We have identified chemical probes that act as dual phosphodiesterase 5 (PDE5) and histone deacetylase 6 (HDAC6)-selective inhibitors (>1 log unit difference versus class I HDACs) to decipher the contribution of HDAC isoforms to the positive impact of dual-acting PDE5 and HDAC inhibitors on mouse models of Alzheimer's disease (AD) and fine-tune this systems therapeutics approach. Structure- and knowledge-based approaches led to the design of first-in-class molecules with the desired target compound profile: dual PDE5 and HDAC6-selective inhibitors. Compound 44b, which fulfilled the biochemical, functional and ADME-Tox profiling requirements and exhibited adequate pharmacokinetic properties, was selected as pharmacological tool compound and tested in a mouse model of AD (Tg2576) in vivo.


Subject(s)
Alzheimer Disease/drug therapy , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Drug Design , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Phosphodiesterase 5 Inhibitors/pharmacology , Alzheimer Disease/metabolism , Cell Line , Dose-Response Relationship, Drug , Histone Deacetylase 6/metabolism , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Humans , Molecular Structure , Neuroglia/drug effects , Phosphodiesterase 5 Inhibitors/chemical synthesis , Phosphodiesterase 5 Inhibitors/chemistry , Structure-Activity Relationship
3.
ACS Chem Neurosci ; 8(3): 638-661, 2017 03 15.
Article in English | MEDLINE | ID: mdl-27936591

ABSTRACT

A novel systems therapeutics approach, involving simultaneous inhibition of phosphodiesterase 5 (PDE5) and histone deacetylase (HDAC), has been validated as a potentially novel therapeutic strategy for the treatment of Alzheimer's disease (AD). First-in-class dual inhibitors bearing a sildenafil core have been very recently reported, and the lead molecule 7 has proven this strategy in AD animal models. Because scaffolds may play a critical role in primary activities and ADME-Tox profiling as well as on intellectual property, we have explored alternative scaffolds (vardenafil- and tadalafil-based cores) and evaluated their impact on critical parameters such as primary activities, permeability, toxicity, and in vivo (pharmacokinetics and functional response in hippocampus) to identify a potential alternative lead molecule bearing a different chemotype for in vivo testing.


Subject(s)
Alzheimer Disease/drug therapy , Histone Deacetylase Inhibitors/therapeutic use , Phosphodiesterase 5 Inhibitors/therapeutic use , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Adenosine Triphosphate/metabolism , Alzheimer Disease/pathology , Animals , Cell Line, Transformed , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Humans , Leukocytes, Mononuclear , Mice , Microsomes, Liver/drug effects , Models, Molecular , Neuroglia/drug effects , Neurons/drug effects , Phosphodiesterase 5 Inhibitors/chemistry , Phosphodiesterase 5 Inhibitors/pharmacology
4.
J Med Chem ; 59(19): 8967-9004, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27606546

ABSTRACT

Simultaneous inhibition of phosphodiesterase 5 (PDE5) and histone deacetylases (HDAC) has recently been validated as a potentially novel therapeutic approach for Alzheimer's disease (AD). To further extend this concept, we designed and synthesized the first chemical series of dual acting PDE5 and HDAC inhibitors, and we validated this systems therapeutics approach. Following the implementation of structure- and knowledge-based approaches, initial hits were designed and were shown to validate our hypothesis of dual in vitro inhibition. Then, an optimization strategy was pursued to obtain a proper tool compound for in vivo testing in AD models. Initial hits were translated into molecules with adequate cellular functional responses (histone acetylation and cAMP/cGMP response element-binding (CREB) phosphorylation in the nanomolar range), an acceptable therapeutic window (>1 log unit), and the ability to cross the blood-brain barrier, leading to the identification of 7 as a candidate for in vivo proof-of-concept testing ( Cuadrado-Tejedor, M.; Garcia-Barroso, C.; Sánchez-Arias, J. A.; Rabal, O.; Mederos, S.; Ugarte, A.; Franco, R.; Segura, V.; Perea, G.; Oyarzabal, J.; Garcia-Osta, A. Neuropsychopharmacology 2016 , in press, doi: 10.1038/npp.2016.163 ).


Subject(s)
Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Phosphodiesterase 5 Inhibitors/chemistry , Phosphodiesterase 5 Inhibitors/pharmacology , Acetylation/drug effects , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Cell Line , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Drug Design , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/pharmacokinetics , Histone Deacetylases/metabolism , Humans , Mice , Models, Molecular , Phosphodiesterase 5 Inhibitors/chemical synthesis , Phosphodiesterase 5 Inhibitors/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL