Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Diego Muñoz-Torrero is active.

Publication


Featured researches published by Diego Muñoz-Torrero.


Current Medicinal Chemistry | 2008

Acetylcholinesterase inhibitors as disease-modifying therapies for Alzheimer's disease.

Diego Muñoz-Torrero

The therapeutic arsenal for the treatment of Alzheimers disease (AD) remains confined to a group of four inhibitors of AChE and one NMDA receptor antagonist, which are used to provide a relief of the very late symptoms of the dementia, i.e. the cognitive and functional decline. In line with the growing body of evidence of the pivotal role of the beta-amyloid peptide (Abeta) in the pathogenesis of AD, alternative classes of drugs targeting mainly the formation or the aggregation of Abeta are actively pursued by the pharmaceutical industry, as they could positively modify the course of AD, stopping or slowing down disease progression. While the first amyloid-directed disease-modifying drugs go ahead with their clinical development and could reach the market as soon as 2009, mounting preclinical and clinical evidences is pointing towards a disease-modifying role also for currently marketed anti-Alzheimer AChE inhibitors (AChEIs), particularly for donepezil. In this review, the neuroprotective effects exhibited by currently commercialized AChEIs will be briefly discussed, together with the secondary mechanisms through which they could exert such effects. This review will focus also on particular classes of AChEIs, namely dual binding site AChEIs, which are being purposely designed to target Abeta aggregation and / or other biological targets that contribute to AD pathogenesis, thus constituting very promising disease-modifying anti-Alzheimer drug candidates.


Journal of Medicinal Chemistry | 2009

Pyrano(3,2-c)quinoline-6-Chlorotacrine Hybrids as a Novel Family of Acetylcholinesterase- and β-Amyloid-Directed Anti-Alzheimer Compounds

Pelayo Camps; Xavier Formosa; Carles Galdeano; Diego Muñoz-Torrero; Lorena Ramírez; Elena Gómez; Nicolas Isambert; Rodolfo Lavilla; Albert Badia; M. Victòria Clos; Manuela Bartolini; Francesca Mancini; Vincenza Andrisano; Mariana P. Arce; M. Isabel Rodríguez-Franco; Oscar Huertas; Thomai Dafni; F. Javier Luque

Two isomeric series of dual binding site acetylcholinesterase (AChE) inhibitors have been designed, synthesized, and tested for their ability to inhibit AChE, butyrylcholinesterase, AChE-induced and self-induced beta-amyloid (Abeta) aggregation, and beta-secretase (BACE-1) and to cross blood-brain barrier. The new hybrids consist of a unit of 6-chlorotacrine and a multicomponent reaction-derived pyrano[3,2-c]quinoline scaffold as the active-site and peripheral-site interacting moieties, respectively, connected through an oligomethylene linker containing an amido group at variable position. Indeed, molecular modeling and kinetic studies have confirmed the dual site binding of these compounds. The new hybrids, and particularly 27, retain the potent and selective human AChE inhibitory activity of the parent 6-chlorotacrine while exhibiting a significant in vitro inhibitory activity toward the AChE-induced and self-induced Abeta aggregation and toward BACE-1, as well as ability to enter the central nervous system, which makes them promising anti-Alzheimer lead compounds.


Current Medicinal Chemistry | 2006

Dimeric and hybrid anti-alzheimer drug candidates

Diego Muñoz-Torrero; Pelayo Camps

In the last decade much attention has been paid to the development of metabolically non-reversible dimeric or hybrid compounds, which combine two structural units of one or two lead compounds of interest for the treatment of Alzheimers disease. As a consequence of their capability to simultaneously interact with two binding sites of the same biological target (the enzyme acetylcholinesterase in most cases), to expand their interaction in the main binding site of the target molecule, or to interact with two different biological targets of interest in the pathogenesis of the disease, these dimeric or hybrid compounds exhibit an improved pharmacological profile including high affinity interactions, additional non conventional actions or complementary actions, what makes them potential drug candidates for the treatment of Alzheimers disease. Herein, we review from a structural point of view the main classes of dimeric or hybrid compounds developed for the treatment of Alzheimers disease, along with the pharmacological profile of the most active compounds.


Bioorganic & Medicinal Chemistry | 1998

Synthesis and evaluation of tacrine–Huperzine a hybrids as acetylcholinesterase inhibitors of potential interest for the treatment of alzheimer’s disease

Albert Badia; Josep Eladi Baños; Pelayo Camps; Joan Picas Contreras; Diana Marina Görbig; Diego Muñoz-Torrero; Montserrat Simon; Nuria M. Vivas

Seventeen polycyclic compounds related to tacrine and huperzine A have been prepared as racemic mixtures and tested as acetylcholinesterase (AChE) inhibitors. The conjunctive pharmacomodulation of huperzine A (carbobicyclic substructure) and tacrine (4-aminoquinoline substructure) led to compound 7jy, 2.5 times less active than tacrine as AChE inhibitor, but much more active than its (Z)-stereoisomer (7iy). Derivatives 7dy and 7ey, lacking the ethylidene substituent, showed to be more active than tacrine. Many other structural modifications of 7jy led to less active compounds. Compounds 7dy and 7ey also showed to be much more active than tacrine in reversing the partial neuromuscular blockade induced by d-tubocurarine.


Journal of Medicinal Chemistry | 2014

Synthesis and multitarget biological profiling of a novel family of rhein derivatives as disease-modifying anti-Alzheimer agents.

Elisabet Viayna; Irene Sola; Manuela Bartolini; Angela De Simone; Cheril Tapia-Rojas; Felipe G. Serrano; Raimon Sabaté; Jordi Juárez-Jiménez; Belén Pérez; F. Javier Luque; Vincenza Andrisano; M. Victòria Clos; Nibaldo C. Inestrosa; Diego Muñoz-Torrero

We have synthesized a family of rhein-huprine hybrids to hit several key targets for Alzheimers disease. Biological screening performed in vitro and in Escherichia coli cells has shown that these hybrids exhibit potent inhibitory activities against human acetylcholinesterase, butyrylcholinesterase, and BACE-1, dual Aβ42 and tau antiaggregating activity, and brain permeability. Ex vivo studies with the leads (+)- and (-)-7e in brain slices of C57bl6 mice have revealed that they efficiently protect against the Aβ-induced synaptic dysfunction, preventing the loss of synaptic proteins and/or have a positive effect on the induction of long-term potentiation. In vivo studies in APP-PS1 transgenic mice treated ip for 4 weeks with (+)- and (-)-7e have shown a central soluble Aβ lowering effect, accompanied by an increase in the levels of mature amyloid precursor protein (APP). Thus, (+)- and (-)-7e emerge as very promising disease-modifying anti-Alzheimer drug candidates.


Journal of Medicinal Chemistry | 2012

Huprine–Tacrine Heterodimers as Anti-Amyloidogenic Compounds of Potential Interest against Alzheimer’s and Prion Diseases

Carles Galdeano; Elisabet Viayna; Irene Sola; Xavier Formosa; Pelayo Camps; Albert Badia; M. Victòria Clos; Júlia Relat; Miriam Ratia; Manuela Bartolini; Francesca Mancini; Vincenza Andrisano; Mario Salmona; Cristina Minguillón; Gema C. González-Muñoz; M. Isabel Rodríguez-Franco; Axel Bidon-Chanal; F. Javier Luque; Diego Muñoz-Torrero

A family of huprine-tacrine heterodimers has been developed to simultaneously block the active and peripheral sites of acetylcholinesterase (AChE). Their dual site binding for AChE, supported by kinetic and molecular modeling studies, results in a highly potent inhibition of the catalytic activity of human AChE and, more importantly, in the in vitro neutralization of the pathological chaperoning effect of AChE toward the aggregation of both the β-amyloid peptide (Aβ) and a prion peptide with a key role in the aggregation of the prion protein. Huprine-tacrine heterodimers take on added value in that they display a potent in vitro inhibitory activity toward human butyrylcholinesterase, self-induced Aβ aggregation, and β-secretase. Finally, they are able to cross the blood-brain barrier, as predicted in an artificial membrane model assay and demonstrated in ex vivo experiments with OF1 mice, reaching their multiple biological targets in the central nervous system. Overall, these compounds are promising lead compounds for the treatment of Alzheimers and prion diseases.


Current Topics in Medicinal Chemistry | 2013

Dual inhibitors of β-amyloid aggregation and acetylcholinesterase as multi-target anti-Alzheimer drug candidates.

Elisabet Viayna; Raimon Sabaté; Diego Muñoz-Torrero

Notwithstanding the functional role that the aggregates of some amyloidogenic proteins can play in different organisms, protein aggregation plays a pivotal role in the pathogenesis of a large number of human diseases. One of such diseases is Alzheimers disease (AD), where the overproduction and aggregation of the β-amyloid peptide (Aβ) are regarded as early critical factors. Another protein that seems to occupy a prominent position within the complex pathological network of AD is the enzyme acetylcholinesterase (AChE), with classical and non-classical activities involved at the late (cholinergic deficit) and early (Aβ aggregation) phases of the disease. Dual inhibitors of Aβ aggregation and AChE are thus emerging as promising multi-target agents with potential to efficiently modify the natural course of AD. In the initial phases of the drug discovery process of such compounds, in vitro evaluation of the inhibition of Aβ aggregation is rather troublesome, as it is very sensitive to experimental assay conditions, and requires expensive synthetic Aβ peptides, which makes cost-prohibitive the screening of large compound libraries. Herein, we review recently developed multitarget anti-Alzheimer compounds that exhibit both Aβ aggregation and AChE inhibitory activities, and, in some cases also additional valuable activities such as BACE-1 inhibition or antioxidant properties. We also discuss the development of simplified in vivo methods for the rapid, simple, reliable, unexpensive, and high-throughput amenable screening of Aβ aggregation inhibitors that rely on the overexpression of Aβ42 alone or fused with reporter proteins in Escherichia coli.


Chemico-Biological Interactions | 2010

Tacrine-based dual binding site acetylcholinesterase inhibitors as potential disease-modifying anti-Alzheimer drug candidates.

Pelayo Camps; Xavier Formosa; Carles Galdeano; Tània Gómez; Diego Muñoz-Torrero; Lorena Ramírez; Elisabet Viayna; Elena Gómez; Nicolas Isambert; Rodolfo Lavilla; Albert Badia; M. Victòria Clos; Manuela Bartolini; Francesca Mancini; Vincenza Andrisano; Axel Bidon-Chanal; Oscar Huertas; Thomai Dafni; F. Javier Luque

Two novel families of dual binding site acetylcholinesterase (AChE) inhibitors have been developed, consisting of a tacrine or 6-chlorotacrine unit as the active site interacting moiety, either the 5,6-dimethoxy-2-[(4-piperidinyl)methyl]-1-indanone fragment of donepezil (or the indane derivative thereof) or a 5-phenylpyrano[3,2-c]quinoline system, reminiscent to the tryciclic core of propidium, as the peripheral site interacting unit, and a linker of suitable length as to allow the simultaneous binding at both sites. These hybrid compounds are all potent and selective inhibitors of human AChE, and more interestingly, are able to interfere in vitro both formation and aggregation of the beta-amyloid peptide, the latter effects endowing these compounds with the potential to modify Alzheimers disease progression.


ChemMedChem | 2010

Novel huprine derivatives with inhibitory activity toward β-amyloid aggregation and formation as disease-modifying anti-Alzheimer drug candidates.

Elisabet Viayna; Tània Gómez; Carles Galdeano; Lorena Ramírez; Miriam Ratia; Albert Badia; M. Victòria Clos; Ester Verdaguer; Felix Junyent; Antoni Camins; Mercè Pallàs; Manuela Bartolini; Francesca Mancini; Vincenza Andrisano; Mariana P. Arce; María Isabel Rodríguez-Franco; Axel Bidon-Chanal; F. Javier Luque; Pelayo Camps; Diego Muñoz-Torrero

A new family of dual binding site acetylcholinesterase (AChE) inhibitors has been designed, synthesized, and tested for their ability to inhibit AChE, butyrylcholinesterase (BChE), AChE‐induced and self‐induced β‐amyloid (Aβ) aggregation and β‐secretase (BACE‐1), and to cross the blood–brain barrier. The new heterodimers consist of a unit of racemic or enantiopure huprine Y or X and a donepezil‐related 5,6‐dimethoxy‐2‐[(4‐piperidinyl)methyl]indane moiety as the active site and peripheral site to mid‐gorge‐interacting moieties, respectively, connected through a short oligomethylene linker. Molecular dynamics simulations and kinetics studies support the dual site binding to AChE. The new heterodimers are potent inhibitors of human AChE and moderately potent inhibitors of human BChE, AChE‐induced and self‐induced Aβ aggregation, and BACE‐1, and are predicted to be able to enter the central nervous system (CNS), thus constituting promising multitarget anti‐Alzheimer drug candidates with the potential to modify the natural course of this disease.


Mini-reviews in Medicinal Chemistry | 2002

Cholinergic Drugs in Pharmacotherapy of Alzheimers Disease

Pelayo Camps; Diego Muñoz-Torrero

The cholinergic hypothesis of Alzheimers disease has spurred the development of numerous structural classes of compounds with different pharmacological profiles aimed at increasing central cholinergic neurotransmission, thus providing a symptomatic treatment for this disease. Indeed, the only drugs currently approved for the treatment of Alzheimers disease are cholinomimetics with the pharmacological profile of acetylcholinesterase inhibitors. Recent evidence of a potential disease modifying role of acetylcholinesterase inhibitors and M(1) muscarinic agonists have led to a revival of this approach, which might be considered as more than a symptomatic treatment.

Collaboration


Dive into the Diego Muñoz-Torrero's collaboration.

Top Co-Authors

Avatar

Pelayo Camps

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Albert Badia

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Belén Pérez

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

M. Victòria Clos

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Irene Sola

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Miriam Ratia

Autonomous University of Barcelona

View shared research outputs
Researchain Logo
Decentralizing Knowledge