Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Eur J Med Chem ; 146: 108-122, 2018 Feb 25.
Article in English | MEDLINE | ID: mdl-29407943

ABSTRACT

In Plasmodium falciparum the bifunctional enzyme glucose-6-phosphate dehydrogenase‒6-phosphogluconolactonase (PfG6PD‒6PGL) is involved in the catalysis of the first reaction of the pentose phosphate pathway. Since this enzyme has a key role in parasite development, its unique structure represents a potential target for the discovery of antimalarial drugs. Here we describe the first 3D structural model of the G6PD domain of PfG6PD‒6PGL. Compared to the human enzyme (hG6PD), the 3D model has enabled the identification of a key difference in the substrate-binding site, which involves the replacement of Arg365 in hG6PD by Asp750 in PfG6PD. In a prospective validation of the model, this critical change has been exploited to rationally design a novel family of substrate analog-based inhibitors that can display the necessary selectivity towards PfG6PD. A series of glucose derivatives featuring an α-methoxy group at the anomeric position and different side chains at position 6 bearing distinct basic functionalities has been synthesized, and their PfG6PD and hG6PD inhibitory activities and their toxicity against parasite and mammalian cells have been assessed. Several compounds displayed micromolar affinity (Ki up to 23 µM), favorable selectivity (up to > 26-fold), and low cytotoxicity. Phenotypic assays with P. falciparum cultures revealed high micromolar IC50 values, likely as a result of poor internalization of the compounds in the parasite cell. Overall, these results endorse confidence to the 3D model of PfG6PD, paving the way for the use of target-based drug design approaches in antimalarial drug discovery studies around this promising target.


Subject(s)
Antimalarials/pharmacology , Drug Discovery , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Plasmodium falciparum/drug effects , Antimalarials/chemical synthesis , Antimalarials/chemistry , Cell Survival/drug effects , Dose-Response Relationship, Drug , Glucosephosphate Dehydrogenase/metabolism , Hep G2 Cells , Humans , Models, Molecular , Molecular Structure , Parasitic Sensitivity Tests , Plasmodium falciparum/cytology , Plasmodium falciparum/enzymology , Structure-Activity Relationship , Tumor Cells, Cultured
2.
Bioorg Med Chem ; 24(21): 5162-5171, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27591008

ABSTRACT

Current drugs against human African trypanosomiasis (HAT) suffer from several serious drawbacks. The search for novel, effective, brain permeable, safe, and inexpensive antitrypanosomal compounds is therefore an urgent need. We have recently reported that the 4-aminoquinoline derivative huprine Y, developed in our group as an anticholinesterasic agent, exhibits a submicromolar potency against Trypanosoma brucei and that its homo- and hetero-dimerization can result in to up to three-fold increased potency and selectivity. As an alternative strategy towards more potent smaller molecule anti-HAT agents, we have explored the introduction of ω-cyanoalkyl, ω-aminoalkyl, or ω-guanidinoalkyl chains at the primary amino group of huprine or the simplified 4-aminoquinoline analogue tacrine. Here, we describe the evaluation of a small in-house library and a second generation of newly synthesized derivatives, which has led to the identification of 13 side chain modified 4-aminoquinoline derivatives with submicromolar potencies against T. brucei. Among these compounds, the guanidinononyltacrine analogue 15e exhibits a 5-fold increased antitrypanosomal potency, 10-fold increased selectivity, and 100-fold decreased anticholinesterasic activity relative to the parent huprine Y. Its biological profile, lower molecular weight relative to dimeric compounds, reduced lipophilicity, and ease of synthesis, make it an interesting anti-HAT lead, amenable to further optimization to eliminate its remaining anticholinesterasic activity.


Subject(s)
Aminoquinolines/pharmacology , Brain/drug effects , Brain/metabolism , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects , Aminoquinolines/chemical synthesis , Aminoquinolines/chemistry , Brain/parasitology , Dose-Response Relationship, Drug , Molecular Structure , Parasitic Sensitivity Tests , Structure-Activity Relationship , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/chemistry
3.
Bioorg Med Chem ; 24(20): 4835-4854, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27396685

ABSTRACT

Different azides and alkynes have been coupled via Cu-catalyzed 1,3-dipolar Huisgen cycloaddition to afford a novel family of N1- and C5-substituted 1,2,3-triazole derivatives that feature the propargylamine group typical of irreversible MAO-B inhibitors at the C4-side chain of the triazole ring. All the synthesized compounds were evaluated against human MAO-A and MAO-B. Structure-activity relationships and molecular modeling were utilized to gain insight into the structural and chemical features that enhance the binding affinity and selectivity between the two enzyme isoforms. Several lead compounds, in terms of potency (submicromolar to low micromolar range), MAO-B selective recognition, and brain permeability, were identified. One of these leads (MAO-B IC50 of 3.54µM, selectivity MAO-A/MAO-B index of 27.7) was further subjected to reversibility and time-dependence inhibition studies, which disclosed a slow and irreversible inhibition of human MAO-B. Overall, the results support the suitability of the 4-triazolylalkyl propargylamine scaffold for exploring the design of multipotent anti-Alzheimer compounds endowed with irreversible MAO-B inhibitory activity.


Subject(s)
Drug Design , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Pargyline/pharmacology , Dose-Response Relationship, Drug , Humans , Molecular Docking Simulation , Molecular Structure , Monoamine Oxidase Inhibitors/chemistry , Pargyline/analogs & derivatives , Pargyline/chemical synthesis , Pargyline/chemistry , Structure-Activity Relationship
4.
Curr Alzheimer Res ; 13(9): 1017-29, 2016.
Article in English | MEDLINE | ID: mdl-26502813

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder in which the amyloid-ß (Aß) peptide plays a key role in synaptic impairment and memory decline associated with neuronal dysfunction and intra-neuronal accumulation of hyperphosphorylated tau protein. Two novel enantiopure rhein-huprine hybrids ((+)-1 and (-)-1) exhibit potent inhibitory effects against human acetylcholinesterase (AChE), butyrylcholinesterase (BuChE), BACE-1 and both Aß and tau antiaggregation activity in vitro and reduction on the amyloid precursor protein (APP) processing in vivo. Interestingly, in this work, we observed beneficial effects with both (+)- and (-)-1 in the reversion of the neuropathology presented in the AßPPswe/PS-1 Alzheimer´s model, including a reduction in the Aß levels, tau phosphorylation and memory impairment with both treatments. Also, in young transgenic mice that present early symptoms of synaptic failure and memory loss, we found a protection of cognitive functions, including long-term potentiation (LTP) and a reduction of the neuro-inflammation by both (+)- and (-)-1. Furthermore, animals with an advanced disease (11month-old) present an exacerbate neurodegeneration that is reversed only with the dextrorotatory enantiomer. These studies indicated that rhein-huprine derivatives with multiple properties might have interesting therapeutic potential for AD.


Subject(s)
Alzheimer Disease/drug therapy , Aminoquinolines/pharmacology , Anthraquinones/pharmacology , Neuroprotective Agents/pharmacology , Nootropic Agents/pharmacology , Aging/drug effects , Aging/metabolism , Aging/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Disease Models, Animal , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Mice, Transgenic , Peptide Fragments/metabolism , Phosphorylation/drug effects , Plaque, Amyloid/drug therapy , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Presenilin-1/genetics , Presenilin-1/metabolism , Synapses/drug effects , Synapses/metabolism , Synapses/pathology , tau Proteins/metabolism
5.
Eur J Med Chem ; 105: 120-37, 2015 Nov 13.
Article in English | MEDLINE | ID: mdl-26479031

ABSTRACT

Human African trypanosomiasis (HAT), Chagas disease and leishmaniasis, which are caused by the trypanosomatids Trypanosoma brucei, Trypanosoma cruzi and Leishmania species, are among the most deadly neglected tropical diseases. The development of drugs that are active against several trypanosomatids is appealing from a clinical and economic viewpoint, and seems feasible, as these parasites share metabolic pathways and hence might be treatable by common drugs. From benzonapthyridine 1, an inhibitor of acetylcholinesterase (AChE) for which we have found a remarkable trypanocidal activity, we have designed and synthesized novel benzo[h][1,6]naphthyridines, pyrrolo[3,2-c]quinolines, azepino[3,2-c]quinolines, and pyrano[3,2-c]quinolines through 2-4-step sequences featuring an initial multicomponent Povarov reaction as the key step. To assess the therapeutic potential of the novel compounds, we have evaluated their in vitro activity against T. brucei, T. cruzi, and Leishmania infantum, as well as their brain permeability, which is of specific interest for the treatment of late-stage HAT. To assess their potential toxicity, we have determined their cytotoxicity against rat myoblast L6 cells and their AChE inhibitory activity. Several tricyclic heterofused quinoline derivatives were found to display an interesting multi-trypanosomatid profile, with one-digit micromolar potencies against two of these parasites and two-digit micromolar potency against the other. Pyranoquinoline 39, which displays IC50 values of 1.5 µM, 6.1 µM and 29.2 µM against T. brucei, L. infantum and T. cruzi, respectively, brain permeability, better drug-like properties (lower lipophilicity and molecular weight and higher CNS MPO desirability score) than hit 1, and the lowest AChE inhibitory activity of the series (IC50 > 30 µM), emerges as an interesting multi-trypanosomatid lead, amenable to further optimization particularly in terms of its selectivity index over mammalian cells.


Subject(s)
Antiprotozoal Agents/chemical synthesis , Antiprotozoal Agents/pharmacology , Leishmania infantum/drug effects , Quinolines/chemical synthesis , Quinolines/pharmacology , Trypanosoma brucei brucei/drug effects , Trypanosoma cruzi/drug effects , Acetylcholinesterase/metabolism , Animals , Antiprotozoal Agents/chemistry , Cell Line , Cell Survival/drug effects , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Electrophorus , Molecular Structure , Parasitic Sensitivity Tests , Quinolines/chemistry , Rats , Structure-Activity Relationship , Trypanosoma brucei brucei/enzymology , Trypanosoma cruzi/enzymology
6.
J Med Chem ; 58(15): 6018-32, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26181606

ABSTRACT

We have synthesized a series of heptamethylene-linked levetiracetam-huprine and levetiracetam-(6-chloro)tacrine hybrids to hit amyloid, tau, and cholinergic pathologies as well as ß-amyloid (Aß)-induced epileptiform activity, some of the mechanisms that eventually lead to cognitive deficits in Alzheimer's disease patients. These hybrids are potent inhibitors of human acetylcholinesterase and butyrylcholinesterase in vitro and moderately potent Aß42 and tau antiaggregating agents in a simple E. coli model of amyloid aggregation. Ex vivo determination of the brain acetylcholinesterase inhibitory activity of these compounds after intraperitoneal injection to C57BL6J mice has demonstrated their ability to enter the brain. The levetiracetam-huprine hybrid 10 significantly reduced the incidence of epileptic seizures, cortical amyloid burden, and neuroinflammation in APP/PS1 mice after a 4-week treatment with a 5 mg/kg dose. Moreover, the hybrid 10 rescued transgenic mice from cognitive deficits, thereby emerging as an interesting disease-modifying anti-Alzheimer drug candidate.


Subject(s)
Alzheimer Disease/drug therapy , Nootropic Agents/pharmacology , Piracetam/analogs & derivatives , Animals , Behavior, Animal/drug effects , In Vitro Techniques , Levetiracetam , Mice , Mice, Inbred C57BL , Nootropic Agents/chemical synthesis , Nootropic Agents/chemistry , Nootropic Agents/therapeutic use , Phenotype , Piracetam/chemical synthesis , Piracetam/chemistry , Piracetam/pharmacology , Piracetam/therapeutic use
7.
Molecules ; 20(3): 4492-515, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25764491

ABSTRACT

We describe the multigram synthesis and in vivo efficacy studies of a donepezil‒huprine hybrid that has been found to display a promising in vitro multitarget profile of interest for the treatment of Alzheimer's disease (AD). Its synthesis features as the key step a novel multigram preparative chromatographic resolution of intermediate racemic huprine Y by chiral HPLC. Administration of this compound to transgenic CL4176 and CL2006 Caenorhabditis elegans strains expressing human Aß42, here used as simplified animal models of AD, led to a significant protection from the toxicity induced by Aß42. However, this protective effect was not accompanied, in CL2006 worms, by a reduction of amyloid deposits. Oral administration for 3 months to transgenic APPSL mice, a well-established animal model of AD, improved short-term memory, but did not alter brain levels of Aß peptides nor cortical and hippocampal amyloid plaque load. Despite the clear protective and cognitive effects of AVCRI104P4, the lack of Aß lowering effect in vivo might be related to its lower in vitro potency toward Aß aggregation and formation as compared with its higher anticholinesterase activities. Further lead optimization in this series should thus focus on improving the anti-amyloid/anticholinesterase activity ratio.


Subject(s)
Alzheimer Disease/drug therapy , Aminoquinolines/administration & dosage , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Indans/administration & dosage , Piperidines/administration & dosage , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Aminoquinolines/chemistry , Aminoquinolines/therapeutic use , Amyloid beta-Protein Precursor/genetics , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Disease Models, Animal , Donepezil , Hep G2 Cells , Heterocyclic Compounds, 4 or More Rings/chemistry , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Humans , Indans/chemistry , Indans/therapeutic use , Mice , Molecular Structure , Piperidines/chemistry , Piperidines/therapeutic use
8.
Bioorg Med Chem ; 23(16): 5156-67, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25678015

ABSTRACT

Dual submicromolar trypanocidal-antiplasmodial compounds have been identified by screening and chemical synthesis of 4-aminoquinoline-based heterodimeric compounds of three different structural classes. In Trypanosoma brucei, inhibition of the enzyme trypanothione reductase seems to be involved in the potent trypanocidal activity of these heterodimers, although it is probably not the main biological target. Regarding antiplasmodial activity, the heterodimers seem to share the mode of action of the antimalarial drug chloroquine, which involves inhibition of the haem detoxification process. Interestingly, all of these heterodimers display good brain permeabilities, thereby being potentially useful for late stage human African trypanosomiasis. Future optimization of these compounds should focus mainly on decreasing cytotoxicity and acetylcholinesterase inhibitory activity.


Subject(s)
Aminoquinolines/chemistry , Aminoquinolines/pharmacology , Antimalarials/chemistry , Antimalarials/pharmacology , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Aminoquinolines/chemical synthesis , Aminoquinolines/pharmacokinetics , Animals , Antimalarials/chemical synthesis , Antimalarials/pharmacokinetics , Brain/metabolism , Cell Line , Dimerization , Hemeproteins/metabolism , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Plasmodium falciparum/drug effects , Plasmodium falciparum/metabolism , Rats , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/pharmacokinetics , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei brucei/enzymology , Trypanosomiasis, African/drug therapy , Trypanosomiasis, African/parasitology
9.
Bioorg Med Chem Lett ; 24(23): 5435-8, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25454267

ABSTRACT

We have synthesized a series of dimers of (+)-(7R,11R)-huprine Y and evaluated their activity against Trypanosoma brucei, Plasmodium falciparum, rat myoblast L6 cells and human acetylcholinesterase (hAChE), and their brain permeability. Most dimers have more potent and selective trypanocidal activity than huprine Y and are brain permeable, but they are devoid of antimalarial activity and remain active against hAChE. Lead optimization will focus on identifying compounds with a more favourable trypanocidal/anticholinesterase activity ratio.


Subject(s)
Antimalarials/pharmacology , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/chemical synthesis , Antiprotozoal Agents/chemistry , Humans , Molecular Structure , Structure-Activity Relationship
10.
J Med Chem ; 57(6): 2549-67, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24568372

ABSTRACT

We have synthesized a family of rhein-huprine hybrids to hit several key targets for Alzheimer's 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 Aß42 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 Aß-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 Aß 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.


Subject(s)
Alzheimer Disease/drug therapy , Anthraquinones/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Protein Precursor/antagonists & inhibitors , Animals , Anthraquinones/pharmacology , Aspartic Acid Endopeptidases/antagonists & inhibitors , Binding Sites , Blood-Brain Barrier/drug effects , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology , Escherichia coli/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Humans , In Vitro Techniques , Kinetics , Long-Term Potentiation/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Molecular , Peptide Fragments/antagonists & inhibitors , Stereoisomerism , Synapses/metabolism , tau Proteins/antagonists & inhibitors
11.
Neurodegener Dis ; 10(1-4): 96-9, 2012.
Article in English | MEDLINE | ID: mdl-22236498

ABSTRACT

BACKGROUND: Multifactorial diseases such as Alzheimer's disease (AD) should be more efficiently tackled by drugs which hit multiple biological targets involved in their pathogenesis. We have recently developed a new family of huprine-tacrine heterodimers, rationally designed to hit multiple targets involved upstream and downstream in the neurotoxic cascade of AD, namely ß-amyloid aggregation and formation as well as acetylcholinesterase catalytic activity. OBJECTIVE: In this study, the aim was to expand the pharmacological profiling of huprine-tacrine heterodimers investigating their effect on muscarinic M(1) receptors as well as their neuroprotective effects against an oxidative insult. METHODS: Sprague-Dawley rat hippocampus homogenates were used to assess the specific binding of two selected compounds in competition with 1 nM [(3)H]pirenzepine (for M(1) receptors) or 0.8 nM [(3)H]quinuclidinyl benzilate (for M(2) receptors). For neuroprotection studies, SHSY5Y cell cultures were subjected to 250 µM hydrogen peroxide insult with or without preincubation with some huprine-tacrine heterodimers. RESULTS: A low nanomolar affinity and M(1)/M(2) selectivity has been found for the selected compounds. Huprine-tacrine heterodimers are not neurotoxic to SHSY5Y cells at a range of concentrations from 1 to 0.001 µM, and some of them can protect cells from the oxidative damage produced by hydrogen peroxide at concentrations as low as 0.001 µM. CONCLUSION: Even though it remains to be determined if these compounds act as agonists at M(1) receptors, as it is the case of the parent huprine Y, their low nanomolar M(1) affinity and neuroprotective effects expand their multitarget profile and increase their interest as disease-modifying anti-Alzheimer agents.


Subject(s)
Aminoquinolines/metabolism , Heterocyclic Compounds, 4 or More Rings/metabolism , Protein Multimerization/physiology , Tacrine/metabolism , Analysis of Variance , Animals , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Hydrogen Peroxide/pharmacology , Muscarinic Antagonists/pharmacokinetics , Neuroblastoma/pathology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Pirenzepine/pharmacokinetics , Protein Binding/drug effects , Protein Multimerization/drug effects , Quinuclidinyl Benzilate/pharmacokinetics , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Tritium/metabolism
12.
J Med Chem ; 55(2): 661-9, 2012 Jan 26.
Article in English | MEDLINE | ID: mdl-22185619

ABSTRACT

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 ß-amyloid peptide (Aß) 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 Aß aggregation, and ß-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 Alzheimer's and prion diseases.


Subject(s)
Alzheimer Disease/drug therapy , Aminoquinolines/chemical synthesis , Amyloid beta-Peptides/antagonists & inhibitors , Cholinesterase Inhibitors/chemical synthesis , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Prion Diseases/drug therapy , Prions/antagonists & inhibitors , Tacrine/analogs & derivatives , Tacrine/chemical synthesis , Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Aminoquinolines/pharmacokinetics , Aminoquinolines/pharmacology , Amyloid beta-Peptides/chemistry , Animals , Brain/metabolism , Butyrylcholinesterase/chemistry , Cholinesterase Inhibitors/pharmacokinetics , Cholinesterase Inhibitors/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Membranes, Artificial , Mice , Models, Molecular , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Permeability , Prions/chemistry , Recombinant Proteins/chemistry , Stereoisomerism , Structure-Activity Relationship , Tacrine/pharmacokinetics , Tacrine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...