Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Nat Rev Drug Discov ; 22(7): 562-584, 2023 07.
Article in English | MEDLINE | ID: mdl-37277503

ABSTRACT

Substitution of a hydrogen atom with its heavy isotope deuterium entails the addition of one neutron to a molecule. Despite being a subtle change, this structural modification, known as deuteration, may improve the pharmacokinetic and/or toxicity profile of drugs, potentially translating into improvements in efficacy and safety compared with the non-deuterated counterparts. Initially, efforts to exploit this potential primarily led to the development of deuterated analogues of marketed drugs through a 'deuterium switch' approach, such as deutetrabenazine, which became the first deuterated drug to receive FDA approval in 2017. In the past few years, the focus has shifted to applying deuteration in novel drug discovery, and the FDA approved the pioneering de novo deuterated drug deucravacitinib in 2022. In this Review, we highlight key milestones in the field of deuteration in drug discovery and development, emphasizing recent and instructive medicinal chemistry programmes and discussing the opportunities and hurdles for drug developers, as well as the questions that remain to be addressed.


Subject(s)
Chemistry, Pharmaceutical , Drug Discovery , Humans , Deuterium/chemistry , Deuterium/pharmacokinetics
2.
Eur J Med Chem ; 252: 115297, 2023 Apr 05.
Article in English | MEDLINE | ID: mdl-36996713

ABSTRACT

Simultaneous modulation of multifaceted toxicity arising from neuroinflammation, oxidative stress, and mitochondrial dysfunction represents a valuable therapeutic strategy to tackle Alzheimer's disease. Among the significant hallmarks of the disorder, Aß protein and its aggregation products are well-recognised triggers of the neurotoxic cascade. In this study, by tailored modification of the curcumin-based lead compound 1, we aimed at developing a small library of hybrid compounds targeting Aß protein oligomerisation and the consequent neurotoxic events. Interestingly, from in vitro studies, analogues 3 and 4, bearing a substituted triazole moiety, emerged as multifunctional agents able to counteract Aß aggregation, neuroinflammation and oxidative stress. In vivo proof-of-concept evaluations, performed in a Drosophila oxidative stress model, allowed us to identify compound 4 as a promising lead candidate.


Subject(s)
Alzheimer Disease , Curcumin , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Curcumin/pharmacology , Curcumin/therapeutic use , Amyloid beta-Peptides/metabolism , Neuroinflammatory Diseases , Oxidative Stress
3.
J Med Chem ; 2022 Nov 02.
Article in English | MEDLINE | ID: mdl-36323630

ABSTRACT

The use of small molecules to induce targeted protein degradation is increasingly growing in the drug discovery landscape, and protein degraders have progressed rapidly through the pipelines. Despite the advances made so far, their synthesis still represents a significant burden and new approaches are highly demanded. Herein we report an unprecedented platform that leverages the modular nature of both multicomponent reactions and degraders to enable the preparation of highly decorated PROTACs. As a proof of principle, our platform has been applied to the preparation of potential BRD4-degrading PROTACs, resulting in the discovery of a set of degraders endowed with high degradation efficiency. Compared to the existing methods, our approach offers a versatile and cost-effective means to access libraries of protein degraders and increase the chance of identifying successful candidates.

4.
Eur J Med Chem ; 241: 114614, 2022 Nov 05.
Article in English | MEDLINE | ID: mdl-35939994

ABSTRACT

Microtubules (MTs) are dynamic filaments of the cytoskeleton, which are formed by the polymerization of their building block tubulin. Perturbation of MT dynamics by MT-targeting agents (MTAs) leads to cell cycle arrest or cell death, a strategy that is pursued in chemotherapy. We recently performed a combined computational and crystallographic fragment screening approach and identified several tubulin-binding fragments. Here, we sought to capitalize on this study with the aim to demonstrate that low affinity tubulin-binding fragments can indeed be used as valuable starting points for the development of active, lead-like antitubulin small molecules. To this end, we report on a new, rationally designed series of 2-aminobenzimidazole derivatives that destabilize MTs by binding tubulin at the colchicine-binding site (CBS). We applied a fragment growing strategy by combining X-ray crystallography and computer-aided drug design. Preliminary structure-activity-relationship studies afforded compound 18 that inhibits HeLa cell viability with a submicromolar activity (IC50 of 0.9 µM). X-ray crystallography confirmed the compound pose in the CBS, while immunostaining experiments suggested a molecular mechanism of action alike classical CBS ligands with antimitotic and antitumor activity associated with MTs destabilization. This promising outcome underpins that our previously performed combined computational and crystallographic fragment screening approach provides promising starting points for developing new MTAs binding to the CBS of tubulin and, eventually, to further tubulin pockets.


Subject(s)
Antineoplastic Agents , Colchicine , Antineoplastic Agents/chemistry , Binding Sites , Cell Proliferation , Colchicine/metabolism , HeLa Cells , Humans , Microtubules/metabolism , Structure-Activity Relationship , Tubulin/metabolism , Tubulin Modulators/chemistry
5.
Int J Mol Sci ; 23(8)2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35457197

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is not restricted to the neuronal compartment but includes important interactions with immune cells, including microglia. Protein aggregates, common pathological hallmarks of AD, bind to pattern recognition receptors on microglia and trigger an inflammatory response, which contributes to disease progression and severity. In this context, curcumin is emerging as a potential drug candidate able to affect multiple key pathways implicated in AD, including neuroinflammation. Therefore, we studied the effect of curcumin and its structurally related analogues cur6 and cur16 on amyloid-ß (Aß)-induced microglia activation and neuronal cell death, as well as their effect on the modulation of Aß aggregation. Primary cortical microglia and neurons were exposed to two different populations of Aß42 oligomers (Aß42Os) where the oligomeric state had been assigned by capillary electrophoresis and ultrafiltration. When stimulated with high molecular weight Aß42Os, microglia released proinflammatory cytokines that led to early neuronal cell death. The studied compounds exerted an anti-inflammatory effect on high molecular weight Aß42O-stimulated microglia and possibly inhibited microglia-mediated neuronal cell toxicity. Furthermore, the tested compounds demonstrated antioligomeric activity during the process of in vitro Aß42 aggregation. These findings could be investigated further and used for the optimization of multipotent candidate molecules for AD treatment.


Subject(s)
Alzheimer Disease , Curcumin , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Cell Death , Curcumin/therapeutic use , Humans , Microglia/metabolism , Peptide Fragments/metabolism
6.
Expert Opin Ther Pat ; 32(6): 605-627, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35235753

ABSTRACT

INTRODUCTION: Compelling evidence identified D3 dopamine receptor (D3R) as a suitable target for therapeutic intervention on CNS-associated disorders, cancer, and other conditions. Several efforts have been made toward developing potent and selective ligands for modulating signaling pathways operated by these GPCRs. The rational design of D3R ligands endowed with a pharmacologically relevant profile has traditionally not encountered much support from computational methods due to a very limited knowledge of the receptor structure and of its conformational dynamics. Recent progress in structural biology will change this state of affairs in the next decade. AREAS COVERED: This review provides an overview of the recent (2014-2020) patent literature on novel classes of D3R ligands developed within the framework of CNS-related diseases, cancer, and additional conditions. When possible, an in-depth description of both in vitro and in vivo generated data is presented. New therapeutic applications of known molecules with activity at D3R are discussed. EXPERT OPINION: Building on current knowledge, future D3R-focused drug discovery campaigns will be propelled by a combination of unprecedented availability of structural information with advanced computational and analytical methods. The design of D3R ligands with the sought activity, efficacy, and selectivity profile will become increasingly more streamlined.


Subject(s)
Patents as Topic , Receptors, Dopamine D3 , Humans , Ligands , Receptors, Dopamine D3/chemistry , Receptors, Dopamine D3/metabolism
7.
Molecules ; 25(13)2020 Jul 05.
Article in English | MEDLINE | ID: mdl-32635622

ABSTRACT

The burden of neoplastic diseases is widely recognized as a severe cause of mortality. The clinical inadequacy of most anticancer therapeutics urgently prompted intense drug discovery efforts toward the identification of new chemical entities endowed with a potent and safe antitumor profile. In this scenario, targeting cancer cells apoptosis machinery has emerged as a relevant strategy, useful for tackling the emergence of drug resistance. On this basis, a small library of naturally inspired hybrid molecules was obtained by combining, through a click chemistry approach, "privileged" synthons such as curcumin scaffold and 1,2,3-triazole building block. Compound 1, bearing a para-fluoro phenyl moiety, showed low-micromolar potency against T acute lymphoblastic leukemia cell growth. More in-depth biologic studies demonstrated, for this analog, cell death-inducing properties associated with its capability to simultaneously activate both the receptor and the mitochondrial apoptosis cascades. This peculiar behavior offers promises for achieving an expanded anticancer effect, namely intense cytotoxic response coupled with reduced predisposition of chemoresistance insurgence. Altogether, this study allowed the identification of compound 1 as a lead compound worth to be progressed as an anticancer drug candidate.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Curcumin/pharmacology , Leukemia, T-Cell/pathology , Membrane Potential, Mitochondrial/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Triazoles/chemistry , Antineoplastic Agents/chemistry , Cell Proliferation , Curcumin/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Leukemia, T-Cell/drug therapy , Molecular Structure , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Structure-Activity Relationship , Tumor Cells, Cultured
8.
ACS Chem Neurosci ; 11(17): 2728-2740, 2020 09 02.
Article in English | MEDLINE | ID: mdl-32663009

ABSTRACT

Common copathogenic factors, including oxidative stress and neuroinflammation, are found to play a vital role in the development of neurodegenerative disorders, including Alzheimer's disease (AD) and Parkinson's disease (PD). Nowadays, owing to the multifactorial character of the diseases, no effective therapies are available, thus underlying the need for new strategies. Overexpression of the enzyme GSK-3ß and downregulation of the Nrf2/ARE pathway are responsible for a decrease in antioxidant defense effects. These pieces of evidence underline the usefulness of dual GSK-3ß inhibitors/Nrf2 inducers. In this regard, to design a dual modulator, the structures of a curcumin-based analogue, as GSK-3ß inhibitor, and a diethyl fumarate fragment, as Nrf2 inducer, were combined. Among the hybrids, 5 and 6 proved to effectively inhibit GSK-3ß, while 4 and 5 showed a marked ability to activate Nrf2 together to increase the neuronal resistance to oxidative stress. These last pieces of evidence translated into specific neuroprotective effects of 4 and 5 against PD pathological events including neurotoxicity elicited by α-synuclein aggregates and 6-hydroxydopamine. Hybrid 5 also showed neuroprotective effects in a C. elegans model of PD where the activation of GSK-3ß is intimately involved in Nrf2 regulation. In summary, 5 emerged as an interesting multitarget derivative, valuable to be exploited in a multitarget PD perspective.


Subject(s)
Curcumin , Parkinson Disease , Animals , Caenorhabditis elegans , Curcumin/pharmacology , Fumarates , Glycogen Synthase Kinase 3 beta , NF-E2-Related Factor 2 , Parkinson Disease/drug therapy
9.
ChemMedChem ; 15(11): 949-954, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32267999

ABSTRACT

Due to the complex and multifactorial nature of bipolar disorder (BD), single-target drugs have traditionally provided limited relief with no disease-modifying effects. In line with the polypharmacology paradigm, we attempted to overcome these limitations by devising two series of multitarget-directed ligands endowed with both a partial agonist profile at dopamine receptor D3 (D3R) and inhibitory activity against glycogen synthase kinase 3 beta (GSK-3ß). These are two structurally unrelated targets that play independent, yet connected, roles in cognition and mood regulation. Two compounds (7 and 10) emerged as promising D3R/GSK-3ß multitarget-directed ligands with nanomolar activity at D3R and low-micromolar inhibition of GSK-3ß, thereby confirming, albeit preliminarily, the feasibility of our strategy. Furthermore, 7 showed promising drug-like properties in stability and pharmacokinetic studies.


Subject(s)
Antipsychotic Agents/pharmacokinetics , Bipolar Disorder/drug therapy , Drug Design , Antipsychotic Agents/chemical synthesis , Antipsychotic Agents/chemistry , Bipolar Disorder/metabolism , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Molecular Structure , Receptors, Dopamine D3/agonists , Receptors, Dopamine D3/metabolism , Structure-Activity Relationship
10.
J Chem Theory Comput ; 15(8): 4646-4659, 2019 Aug 13.
Article in English | MEDLINE | ID: mdl-31246463

ABSTRACT

It is widely accepted that drug-target association and dissociation rates directly affect drug efficacy and safety. To rationally optimize drug binding kinetics, one must know the atomic arrangement of the protein-ligand complex during the binding/unbinding process in order to detect stable and metastable states. Whereas experimental approaches can determine kinetic constants with fairly good accuracy, computational approaches based on molecular dynamics (MD) simulations can deliver the atomistic details of the unbinding process. Furthermore, they can also be utilized prospectively to predict residence time (i.e., the inverse of unbinding kinetics constant, koff) with an acceptable level of accuracy. Here, we report a novel method based on adiabatic bias MD with an electrostatics-like collective variable (dubbed elABMD) for sampling protein-ligand dissociation events in two kinases. elABMD correctly ranked a ligand series on glucokinase, in agreement with experimental data and previous calculations. Subsequently, we applied the new method prospectively to a congeneric series of GSK-3ß inhibitors. For this series, new crystal structures were generated and the residence time was experimentally measured with surface plasmon resonance (SPR). There was good agreement between computational predictions and experimental measures, suggesting that elABMD is an innovative and efficient tool for calculating residence times.


Subject(s)
Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/metabolism , Protein Kinase Inhibitors/pharmacology , Crystallography, X-Ray , Glycogen Synthase Kinase 3 beta/chemistry , Humans , Kinetics , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Protein Kinase Inhibitors/chemistry , Static Electricity , Thermodynamics
11.
Expert Opin Ther Pat ; 27(8): 953-965, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28597702

ABSTRACT

INTRODUCTION: Curcumin, the main bioactive compound found in the rhizome of Curcuma longa L., is considered a 'privileged structure', due to its ability to modulate different signaling pathways involved in the pathogenesis of several diseases. Unfortunately, its poor pharmacodynamic and pharmacokinetic properties, mainly related to chemical instability, low solubility and rapid metabolism, greatly reduce its therapeutic potential. In the last years a number of derivatives were developed and patented, aimed both at improving its multifaceted biological profile and overcoming its undesired effects. Areas covered: This review summarizes the patent literature of the last five years dealing with synthetic curcumin-related compounds in cancer and neurodegeneration, properly designed in order to avoid the so-called 'dark side of curcumin', and to take advantage of the beneficial properties of this molecule, worth to be further exploited to obtain effective therapeutics. Expert opinion: Due to the synergistic binding to several networked targets, curcumin turned out to be suitable for polypharmacological approaches, and its 'privileged structure' could also provide the key scaffold to develop novel multipotent drugs useful for treating multifactiorial pathologic conditions such as cancer and neurodegeneration.


Subject(s)
Antineoplastic Agents/administration & dosage , Curcumin/administration & dosage , Drug Design , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Curcuma/chemistry , Curcumin/chemistry , Curcumin/pharmacokinetics , Drug Synergism , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/physiopathology , Patents as Topic , Solubility
12.
Br J Pharmacol ; 174(10): 1090-1103, 2017 05.
Article in English | MEDLINE | ID: mdl-28198010

ABSTRACT

BACKGROUND AND PURPOSE: Toll-like receptor 4 (TLR4) plays a key role in the induction of inflammatory responses both in peripheral organs and the CNS. Curcumin exerts anti-inflammatory functions by interfering with LPS-induced dimerization of TLR4-myeloid differentiation protein-2 (MD-2) complex and suppressing pro-inflammatory mediator release. However, the inhibitory mechanism of curcumin remains to be defined. EXPERIMENTAL APPROACH: Binding of bis-demethoxycurcumin (GG6) and its cyclized pyrazole analogue (GG9), lacking the 1,3-dicarbonyl function, to TLR4-MD-2 was determined using molecular docking simulations. The effects of these compounds on cytokine release and NF-κB activation were examined by ELISA and fluorescence staining in LPS-stimulated primary microglia. Interference with TLR4 dimerization was assessed by immunoprecipitation in Ba/F3 cells. KEY RESULTS: Both curcumin analogues bound to the hydrophobic region of the MD-2 pocket. However, only curcumin and GG6, both possessing the 1,3-diketone moiety, inhibited LPS-induced TLR4 dimerization, activation of NF-κB and secretion of pro-inflammatory cytokines in primary microglia. Consistent with the ability of 1,3-diketones to coordinate divalent metal ions, LPS stimulation in a low magnesium environment decreased pro-inflammatory cytokine release and NF-κB p65 nuclear translocation in microglia and decreased TLR4-MD-2 dimerization in Ba/F3 cells. Curcumin and GG6 also significantly reduced cytokine output in contrast to the pyrazole analogue GG9. CONCLUSIONS AND IMPLICATIONS: These results indicate that phenolic 1,3-diketones, with a structural motif able to coordinate magnesium ions, can modulate LPS-mediated TLR4-MD-2 signalling. Taken together, these studies identify a previously uncharacterized mechanism involving magnesium, underlying the inflammatory responses to LPS.


Subject(s)
Inflammation/drug therapy , Ketones/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Magnesium/metabolism , Animals , Cells, Cultured , Cytokines/metabolism , Dose-Response Relationship, Drug , Female , Inflammation/metabolism , Ketones/chemistry , Lipopolysaccharides/pharmacology , Lymphocyte Antigen 96/antagonists & inhibitors , Lymphocyte Antigen 96/metabolism , Male , Microglia/drug effects , Microglia/metabolism , Molecular Structure , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Structure-Activity Relationship , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/metabolism
13.
J Med Chem ; 60(6): 2287-2304, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28182408

ABSTRACT

We recently reported molecules designed according to the multitarget-directed ligand paradigm to exert combined activity at human fatty acid amide hydrolase (FAAH) and dopamine receptor subtype D3 (D3R). Both targets are relevant for tackling several types of addiction (most notably nicotine addiction) and other compulsive behaviors. Here, we report an SAR exploration of a series of biphenyl-N-[4-[4-(2,3-substituted-phenyl)piperazine-1-yl]alkyl]carbamates, a novel class of molecules that had shown promising activities at the FAAH-D3R target combination in preliminary studies. We have rationalized the structural features conducive to activities at the main targets and investigated activities at two off-targets: dopamine receptor subtype D2 and endocannabinoid receptor CB1. To understand the unexpected affinity for the CB1 receptor, we devised a 3D-QSAR model, which we then prospectively validated. Compound 33 was selected for PK studies because it displayed balanced affinities for the main targets and clear selectivity over the two off-targets. 33 has good stability and oral bioavailability and can cross the blood-brain barrier.


Subject(s)
Amidohydrolases/metabolism , Carbamates/chemistry , Carbamates/pharmacology , Drug Design , Piperazines/chemistry , Piperazines/pharmacology , Receptors, Dopamine D3/metabolism , Amidohydrolases/antagonists & inhibitors , Animals , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacokinetics , Biphenyl Compounds/pharmacology , Blood-Brain Barrier/metabolism , CHO Cells , Carbamates/chemical synthesis , Carbamates/pharmacokinetics , Cricetulus , HEK293 Cells , Humans , Male , Models, Molecular , Piperazine , Piperazines/chemical synthesis , Piperazines/pharmacokinetics , Quantitative Structure-Activity Relationship , Rats, Sprague-Dawley , Receptors, Dopamine D3/agonists , Receptors, Dopamine D3/antagonists & inhibitors
14.
Expert Opin Ther Pat ; 27(5): 579-590, 2017 May.
Article in English | MEDLINE | ID: mdl-28024125

ABSTRACT

INTRODUCTION: curcumin is the main bioactive component contained in Curcuma Longa, largely employed in traditional medicine. Recently, beneficial properties, useful for prevention and treatment of several disorders, have been discovered for this compound. Peculiar structural feature is an α,ß-unsaturated carbonyl system essential for establishing contacts with critical cysteine residues of several targets. This distinctive mechanism of action imparts to the molecule the ability to affect a large number of targets, accounting for its pleiotropic behaviour and definition of "privileged structure". Areas covered: The objective of the review is an examination of the recent developments in the field of the anti-cancer applications of curcumin, together with formulation issues, considering the patent literature in the years 2012-2016. Expert opinion: The wide therapeutic efficacy of curcumin is related to synergistic interactions with several biological targets, along with the modulation of several signaling pathways. This peculiar behaviour could be useful in the treatment of multifactorial diseases such as cancer. Combination of curcumin with a first line antineoplastic drug proved to be a valuable strategy to obtain an amplified response with minimized side effects. Innovative curcumin formulations based on the nanotechnology approach allowed improving both bioavailability and therapeutic efficacy.


Subject(s)
Antineoplastic Agents/administration & dosage , Curcumin/administration & dosage , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Biological Availability , Curcuma/chemistry , Curcumin/pharmacokinetics , Curcumin/therapeutic use , Drug Design , Humans , Nanotechnology , Patents as Topic
15.
Molecules ; 21(5)2016 May 16.
Article in English | MEDLINE | ID: mdl-27196880

ABSTRACT

Alzheimer's disease (AD) has been defined as a multi-factorial disorder resulting from a complex array of networked cellular and molecular mechanisms. In particular, elevated levels of Aß protein and its aggregation products in the presence of metal ions proved to be highly neurotoxic and therapeutic strategies aimed at preventing Aß generation and oxidative stress may represent an effective approach for AD treatment. A recent paradigm for the treatment of complex diseases such as AD suggests the employment of multifunctional compounds, single chemical entities capable of simultaneously modulating different targets involved in the pathology. In this paper, the "pharmacophores combination" strategy was applied, connecting the main scaffold of the BACE-1 ligand 1 to that of the chalcone 2, as metal chelating pharmacophore, to obtain a small library of compounds. Conjugate 5 emerged as the most interesting derivative, proving to inhibit BACE-1 with low-micromolar potency, and showing neuroprotective effects. In particular, 5 proved to be able to protect from metal-associated oxidative stress by hampering intracellular Cu(2+)-induced ROS formation without any direct neurotoxic effect.


Subject(s)
Alzheimer Disease/drug therapy , Chalcone/administration & dosage , Neuroprotective Agents/administration & dosage , Protein Aggregation, Pathological/drug therapy , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/chemistry , Amyloid beta-Peptides/chemistry , Aspartic Acid Endopeptidases/chemistry , Chalcone/chemistry , Chelating Agents/administration & dosage , Chelating Agents/chemistry , Drug Combinations , Humans , Ions/chemistry , Ligands , Metals/chemistry , Neuroprotective Agents/chemistry , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
16.
J Med Chem ; 59(2): 531-44, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26696252

ABSTRACT

The multitarget approach has gained increasing acceptance as a useful tool to address complex and multifactorial maladies such as Alzheimer's disease (AD). The concurrent inhibition of the validated AD targets ß-secretase (BACE-1) and glycogen synthase kinase-3ß (GSK-3ß) by attacking both ß-amyloid and tau protein cascades has been identified as a promising AD therapeutic strategy. In our study, curcumin was identified as a lead compound for the simultaneous inhibition of both targets; therefore, synthetic efforts were dedicated to obtaining a small library of novel curcumin-based analogues, and a number of potent and balanced dual-target inhibitors were obtained. In particular, 2, 6, and 7 emerged as promising drug candidates endowed with neuroprotective potential and brain permeability. Notably, for some new compounds the symmetrical diketo and the ß-keto-enol tautomeric forms were purposely isolated and tested in vitro, allowing us to gain insight into the key requirements for BACE-1 and GSK-3ß inhibition.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Curcumin/chemistry , Curcumin/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Blood-Brain Barrier , Brain/metabolism , Cell Line, Tumor , Cell Survival , Drug Design , Enzyme Induction/drug effects , Glycogen Synthase Kinase 3 beta , Humans , Models, Molecular , Molecular Docking Simulation , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , Neuroprotective Agents/pharmacology , Small Molecule Libraries , Structure-Activity Relationship
17.
Bioorg Med Chem Lett ; 25(14): 2804-8, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26003339

ABSTRACT

In recent years, a progressive increase in age-related disorders could be observed in most western countries, among which Alzheimer's disease (AD) is one of the most challenging. BACE1 could be seen as an attractive target to develop disease-modifying compounds, and in this context, a new series of hybrid molecules was designed and synthesized, based on a previously identified multitarget lead compound. In particular, the amino side chain was appropriately modified to fit BACE1 as additional target. In vitro testing results pointed out compound 8 (IC50=2.49±0.08 µM), bearing the bulky bis(4-fluorophenyl)methyl)piperazine substituent, as the most potent BACE1 inhibitor of the series.


Subject(s)
Acetylcholinesterase/chemistry , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Cholinesterase Inhibitors/chemistry , Indans/chemistry , Protease Inhibitors/chemistry , Acetylcholinesterase/metabolism , Amines/chemistry , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Binding Sites , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/metabolism , Drug Design , Humans , Indans/chemical synthesis , Indans/metabolism , Molecular Docking Simulation , Protease Inhibitors/chemical synthesis , Protease Inhibitors/metabolism , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...