Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 273
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Pharm Res ; 41(5): 863-875, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38605261

RESUMO

OBJECTIVE: This study aimed to improve the efficiency of pharmacotherapy for CNS diseases by optimizing the ability of drug molecules to penetrate the Blood-Brain Barrier (BBB). METHODS: We established qualitative and quantitative databases of the ADME properties of drugs and derived characteristic features of compounds with efficient BBB penetration. Using these insights, we developed four machine learning models to predict a drug's BBB permeability by assessing ADME properties and molecular topology. We then validated the models using the B3DB database. For acyclovir and ceftriaxone, we modified the Hydrogen Bond Donors and Acceptors, and evaluated the BBB permeability using the predictive model. RESULTS: The machine learning models performed well in predicting BBB permeability on both internal and external validation sets. Reducing the number of Hydrogen Bond Donors and Acceptors generally improves BBB permeability. Modification only enhanced BBB penetration in the case of acyclovir and not ceftriaxone. CONCLUSIONS: The machine learning models developed can accurately predict BBB permeability, and many drug molecules are likely to have increased BBB penetration if the number of Hydrogen Bond Donors and Acceptors are reduced. These findings suggest that molecular modifications can enhance the efficacy of CNS drugs and provide practical strategies for drug design and development. This is particularly relevant for improving drug penetration of the BBB.


Assuntos
Aciclovir , Barreira Hematoencefálica , Aprendizado de Máquina , Permeabilidade , Barreira Hematoencefálica/metabolismo , Humanos , Aciclovir/farmacocinética , Ligação de Hidrogênio , Ceftriaxona/farmacocinética , Fármacos do Sistema Nervoso Central/farmacocinética , Fármacos do Sistema Nervoso Central/química , Fármacos do Sistema Nervoso Central/metabolismo , Desenho de Fármacos
2.
Proc Natl Acad Sci U S A ; 117(51): 32691-32700, 2020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33288687

RESUMO

Preclinical and clinical data support the use of focused ultrasound (FUS), in the presence of intravenously injected microbubbles, to safely and transiently increase the permeability of the blood-brain barrier (BBB). FUS-induced BBB permeability has been shown to enhance the bioavailability of administered intravenous therapeutics to the brain. Ideal therapeutics candidates for this mode of delivery are those capable of inducing benefits peripherally following intravenous injection and in the brain at FUS-targeted areas. In Alzheimer's disease, intravenous immunoglobulin (IVIg), a fractionated human blood product containing polyclonal antibodies, act as immunomodulator peripherally and centrally, and it can reduce amyloid pathology in the brain. Using the TgCRND8 mouse model of amyloidosis, we tested whether FUS can improve the delivery of IVIg, administered intravenously (0.4 g/kg), to the hippocampus and reach an effective dose to reduce amyloid plaque pathology and promote neurogenesis. Our results show that FUS-induced BBB permeability is required to deliver a significant amount of IVIg (489 ng/mg) to the targeted hippocampus of TgCRN8 mice. Two IVIg-FUS treatments, administered at days 1 and 8, significantly increased hippocampal neurogenesis by 4-, 3-, and 1.5-fold in comparison to saline, IVIg alone, and FUS alone, respectively. Amyloid plaque pathology was significantly reduced in all treatment groups: IVIg alone, FUS alone, and IVIg-FUS. Putative factors promoting neurogenesis in response to IVIg-FUS include the down-regulation of the proinflammatory cytokine TNF-α in the hippocampus. In summary, FUS was required to deliver an effective dose of IVIg to promote hippocampal neurogenesis and modulate the inflammatory milieu.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Sistemas de Liberação de Medicamentos/métodos , Hipocampo/efeitos dos fármacos , Imunoglobulinas Intravenosas/administração & dosagem , Imunoglobulinas Intravenosas/farmacologia , Ultrassom/métodos , Doença de Alzheimer/patologia , Animais , Disponibilidade Biológica , Barreira Hematoencefálica/efeitos dos fármacos , Fármacos do Sistema Nervoso Central/administração & dosagem , Fármacos do Sistema Nervoso Central/farmacocinética , Modelos Animais de Doenças , Feminino , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Imunoglobulinas Intravenosas/farmacocinética , Imageamento por Ressonância Magnética , Masculino , Camundongos Transgênicos , Microbolhas , Neurogênese/efeitos dos fármacos , Neurogênese/fisiologia , Placa Amiloide/tratamento farmacológico , Placa Amiloide/patologia , Resultado do Tratamento , Fator de Necrose Tumoral alfa/metabolismo
3.
J Chem Inf Model ; 62(11): 2685-2695, 2022 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-35581002

RESUMO

The aim of drug design and development is to produce a drug that can inhibit the target protein and possess a balanced physicochemical and toxicity profile. Traditionally, this is a multistep process where different parameters such as activity and physicochemical and pharmacokinetic properties are optimized sequentially, which often leads to high attrition rate during later stages of drug design and development. We have developed a deep learning-based de novo drug design method that can design novel small molecules by optimizing target specificity as well as multiple parameters (including late-stage parameters) in a single step. All possible combinations of parameters were optimized to understand the effect of each parameter over the other parameters. An explainable predictive model was used to identify the molecular fragments responsible for the property being optimized. The proposed method was applied against the human 5-hydroxy tryptamine receptor 1B (5-HT1B), a protein from the central nervous system (CNS). Various physicochemical properties specific to CNS drugs were considered along with the target specificity and blood-brain barrier permeability (BBBP), which act as an additional challenge for CNS drug delivery. The contribution of each parameter toward molecule design was identified by analyzing the properties of generated small molecules from optimization of all possible parameter combinations. The final optimized generative model was able to design similar inhibitors compared to known inhibitors of 5-HT1B. In addition, the functional groups of the generated small molecules that guide the BBBP predictive model were identified through feature attribution techniques.


Assuntos
Sistema Nervoso Central , Desenho de Fármacos , Barreira Hematoencefálica/metabolismo , Sistema Nervoso Central/metabolismo , Fármacos do Sistema Nervoso Central/química , Fármacos do Sistema Nervoso Central/farmacocinética , Humanos , Preparações Farmacêuticas/metabolismo
4.
Handb Exp Pharmacol ; 273: 121-150, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33258066

RESUMO

It is crucial to understand the basic principles of drug transport, from the site of delivery to the site of action within the CNS, in order to evaluate the possible utility of a new drug candidate for CNS action, or possible CNS side effects of non-CNS targeting drugs. This includes pharmacokinetic aspects of drug concentration-time profiles in plasma and brain, blood-brain barrier transport and drug distribution within the brain parenchyma as well as elimination processes from the brain. Knowledge of anatomical and physiological aspects connected with drug delivery is crucial in this context. The chapter is intended for professionals working in the field of CNS drug development and summarizes key pharmacokinetic principles and state-of-the-art experimental methodologies to assess brain drug disposition. Key parameters, describing the extent of unbound (free) drug across brain barriers, in particular blood-brain and blood-cerebrospinal fluid barriers, are presented along with their application in drug development. Special emphasis is given to brain intracellular pharmacokinetics and its role in evaluating target engagement. Fundamental neuropharmacokinetic differences between small molecular drugs and biologicals are discussed and critical knowledge gaps are outlined.


Assuntos
Barreira Hematoencefálica , Encéfalo , Transporte Biológico/fisiologia , Fármacos do Sistema Nervoso Central/farmacocinética , Humanos , Preparações Farmacêuticas
5.
Drug Metab Dispos ; 49(1): 3-11, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33144341

RESUMO

Understanding the mechanisms of drug transport across the blood-brain barrier (BBB) is an important issue for regulating the pharmacokinetics of drugs in the central nervous system. In this study, we focused on solute carrier family 35, member F2 (SLC35F2), whose mRNA is highly expressed in the BBB. SLC35F2 protein was enriched in isolated mouse and monkey brain capillaries relative to brain homogenates and was localized exclusively on the apical membrane of MDCKII cells and brain microvascular endothelial cells (BMECs) differentiated from human induced pluripotent stem cells (hiPS-BMECs). SLC35F2 activity was assessed using its substrate, YM155, and pharmacological experiments revealed SLC35F2 inhibitors, such as famotidine (half-maximal inhibitory concentration, 160 µM). Uptake of YM155 was decreased by famotidine or SLC35F2 knockdown in immortalized human BMECs (human cerebral microvascular endothelial cell/D3 cells). Furthermore, famotidine significantly inhibited the apical (A)-to-basal (B) transport of YM155 in primary cultured monkey BMECs and hiPS-BMECs. Crucially, SLC35F2 knockout diminished the A-to-B transport and intracellular accumulation of YM155 in hiPS-BMECs. By contrast, in studies using an in situ brain perfusion technique, neither deletion of Slc35f2 nor famotidine reduced brain uptake of YM155, even though YM155 is a substrate of mouse SLC35F2. YM155 uptake was decreased significantly by losartan and naringin, inhibitors for the organic anion transporting polypeptide (OATP) 1A4. These findings suggest SLC35F2 is a functional transporter in various cellular models of the primate BBB that delivers its substrates to the brain and that its relative importance in the BBB is modified by differences in the expression of OATPs between primates and rodents. SIGNIFICANCE STATEMENT: This study demonstrated that SLC35F2 is a functional drug influx transporter in three different cellular models of the primate blood-brain barrier (i.e., human cerebral microvascular endothelial cell/D3 cells, primary cultured monkey BMECs, and human induced pluripotent stem-BMECs) but has limited roles in mouse brain. SLC35F2 facilitates apical-to-basal transport across the tight cell monolayer. These findings will contribute to the development of improved strategies for targeting drugs to the central nervous system.


Assuntos
Transporte Biológico/efeitos dos fármacos , Barreira Hematoencefálica , Famotidina/farmacocinética , Imidazóis/farmacocinética , Proteínas de Membrana Transportadoras/metabolismo , Naftoquinonas/farmacocinética , Transportadores de Ânions Orgânicos/metabolismo , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Células Cultivadas , Fármacos do Sistema Nervoso Central/farmacocinética , Desenvolvimento de Medicamentos/métodos , Células Endoteliais/metabolismo , Haplorrinos , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Modelos Biológicos
6.
Pharmacol Res ; 166: 105480, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33549730

RESUMO

Lithium (Li) is the most widely used mood stabilizer in treating patients with bipolar disorder. However, more than half of the patients do not or partially respond to Li therapy, despite serum Li concentrations in the serum therapeutic range. The exact mechanisms underlying the pharmacokinetic-pharmacodynamic (PK-PD) relationships of lithium are still poorly understood and alteration in the brain pharmacokinetics of lithium may be one of the mechanisms explaining the variability in the clinical response to Li. Brain barriers such as the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB) play a crucial role in controlling blood-to-brain and brain-to-blood exchanges of various molecules including central nervous system (CNS) drugs. Recent in vivo studies by nuclear resonance spectroscopy revealed heterogenous brain distribution of Li in human that were not always correlated with serum concentrations, suggesting regional and variable transport mechanisms of Li through the brain barriers. Moreover, alteration in the functionality and integrity of brain barriers is reported in various CNS diseases, as a cause or a consequence and in this regard, Li by itself is known to modulate BBB properties such as the expression and activity of various transporters, metabolizing enzymes, and the specialized tight junction proteins on BBB. In this review, we will focus on recent knowledge into the role of the brain barriers as key-element in the Li neuropharmacokinetics which might improve the understanding of PK-PD of Li and its interindividual variability in drug response.


Assuntos
Transtorno Bipolar/tratamento farmacológico , Encéfalo/metabolismo , Fármacos do Sistema Nervoso Central/farmacocinética , Compostos de Lítio/farmacocinética , Animais , Antimaníacos/farmacocinética , Antimaníacos/farmacologia , Antimaníacos/uso terapêutico , Transporte Biológico/efeitos dos fármacos , Transtorno Bipolar/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Encéfalo/efeitos dos fármacos , Fármacos do Sistema Nervoso Central/farmacologia , Fármacos do Sistema Nervoso Central/uso terapêutico , Humanos , Compostos de Lítio/farmacologia , Compostos de Lítio/uso terapêutico
7.
Bull Math Biol ; 81(9): 3508-3541, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-29230702

RESUMO

Positron emission tomography, an imaging tool using radiolabeled tracers in humans and preclinical species, has been widely used in recent years in drug development, particularly in the central nervous system. One important goal of PET in drug development is assessing the occupancy of various molecular targets (e.g., receptors, transporters, enzymes) by exogenous drugs. The current linear mathematical approaches used to determine occupancy using PET imaging experiments are presented. These algorithms use results from multiple regions with different target content in two scans, a baseline (pre-drug) scan and a post-drug scan. New mathematical estimation approaches to determine target occupancy, using maximum likelihood, are presented. A major challenge in these methods is the proper definition of the covariance matrix of the regional binding measures, accounting for different variance of the individual regional measures and their nonzero covariance, factors that have been ignored by conventional methods. The novel methods are compared to standard methods using simulation and real human occupancy data. The simulation data showed the expected reduction in variance and bias using the proper maximum likelihood methods, when the assumptions of the estimation method matched those in simulation. Between-method differences for data from human occupancy studies were less obvious, in part due to small dataset sizes. These maximum likelihood methods form the basis for development of improved PET covariance models, in order to minimize bias and variance in PET occupancy studies.


Assuntos
Desenvolvimento de Medicamentos/métodos , Modelos Biológicos , Tomografia por Emissão de Pósitrons/métodos , Animais , Sítios de Ligação , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Fármacos do Sistema Nervoso Central/farmacocinética , Simulação por Computador , Desenvolvimento de Medicamentos/estatística & dados numéricos , Humanos , Funções Verossimilhança , Conceitos Matemáticos , Modelos Neurológicos , Farmacocinética , Tomografia por Emissão de Pósitrons/estatística & dados numéricos , Receptores de Droga/metabolismo , Receptores Opioides kappa/metabolismo
8.
Biol Pharm Bull ; 41(9): 1330-1336, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30175770

RESUMO

Nicotine, an addictive substance, is absorbed from the lungs following inhalation of tobacco smoke, and distributed to various tissues such as liver, brain, and retina. Recent in vivo and in vitro studies suggest the involvement of a carrier-mediated transport process in nicotine transport in the lung, liver, and inner blood-retinal barrier. In addition, in vivo studies of influx and efflux transport of nicotine across the blood-brain barrier (BBB) revealed that blood-to-brain influx transport of nicotine is more dominant than brain-to-blood efflux transport of nicotine. Uptake studies in TR-BBB13 cells, which are an in vitro model cell line of the BBB, suggest the involvement of H+/organic cation antiporter, which is distinct from typical organic cation transporters, in nicotine transport at the BBB. Moreover, inhibition studies in TR-BBB13 cells showed that nicotine uptake was significantly reduced by central nervous system (CNS) drugs, such as antidepressants, anti-Alzheimer's disease drugs, and anti-Parkinson's disease drugs, suggesting that the nicotine transport system can recognize these molecules. The cumulative evidence would be helpful to improve our understanding of smoking-CNS drug interaction for providing appropriate medication.


Assuntos
Barreira Hematoencefálica/metabolismo , Fármacos do Sistema Nervoso Central/farmacocinética , Nicotina/farmacocinética , Animais , Transporte Biológico , Interações Medicamentosas , Humanos
9.
Molecules ; 23(6)2018 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-29843371

RESUMO

Background: The blood brain barrier (BBB) is a dynamic and functional structure which poses a vast challenge in the development of drugs acting on the central nervous system (CNS). While most substances are denied BBB crossing, selective penetration of substances mainly occurs through diffusion, carrier mediated transport, or receptor mediated transcytosis. Methods: Strategies in enhancing BBB penetration have been reviewed and summarized in accordance with their type of formulation. Highlights in monoclonal antibodies, peptide-vectors, nanoparticles, and simple prodrugs were included. Conclusion: Nanoparticles and simple prodrugs, for example, can be used for efficient BBB penetration through inhibition of efflux mechanisms, however, monoclonal antibodies are the most promising strategy in BBB penetration. Close follow-up of future development in this area should confirm our expectation.


Assuntos
Barreira Hematoencefálica/química , Fármacos do Sistema Nervoso Central/farmacocinética , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacocinética , Transporte Biológico , Fármacos do Sistema Nervoso Central/química , Humanos , Estrutura Molecular , Nanoestruturas/química , Pró-Fármacos/química , Pró-Fármacos/farmacocinética
10.
J Neurochem ; 140(5): 692-702, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27925201

RESUMO

Angiogenesis and inflammation are clearly interconnected and interdependent processes that are dysregulated in a series of systemic and brain pathologies. Herein, key aspects regarding endothelial cell function and tissue remodelling that are particularly affected or aggravated by inflammation are presented. Most importantly, the cellular and molecular mechanisms involved in the vascular regulation of the inflammatory processes occurring in several brain disorders and how they impact on disease/injury progression are detailed, highlighting potential targets for therapy. Finally, nanomedicine-based approaches designed to overcome limitations pertaining to low systemic bioavailability, light, pH and temperature sensitivity and/or rapid degradation of these targets, and to optimize their mode of action are discussed. Ultimately, we expect this review to provide new insight and to suggest novel approaches for the treatment of blood-brain barrier dysfunction per se or as a means to treat the injured or diseased central nervous system.


Assuntos
Vasos Sanguíneos/fisiopatologia , Fármacos do Sistema Nervoso Central/uso terapêutico , Inflamação/fisiopatologia , Animais , Encefalopatias/tratamento farmacológico , Fármacos do Sistema Nervoso Central/farmacocinética , Endotélio Vascular/efeitos dos fármacos , Humanos , Neovascularização Fisiológica/efeitos dos fármacos
11.
Alzheimers Dement ; 13(4): 419-430, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27693181

RESUMO

INTRODUCTION: Amyloid imaging has been integrated into diagnostic criteria for Alzheimer's disease (AD). How amyloid tracers binding differ for different tracer structures and amyloid-ß aggregates in autosomal dominant AD (ADAD) and sporadic AD is unclear. METHODS: Binding properties of different amyloid tracers were examined in brain homogenates from six ADAD with APPswe, PS1 M146V, and PS1 EΔ9 mutations, 13 sporadic AD, and 14 control cases. RESULTS: 3H-PIB, 3H-florbetaben, 3H-AZD2184, and BTA-1 shared a high- and a varying low-affinity binding site in the frontal cortex of sporadic AD. AZD2184 detected another binding site (affinity 33 nM) in the frontal cortex of ADAD. The 3H-AZD2184 and 3H-PIB binding were significantly higher in the striatum of ADAD compared to sporadic AD and control. Polyphenol resveratrol showed strongest inhibition on 3H-AZD84 binding followed by 3H-florbetaben and minimal on 3H-PIB. DISCUSSION: This study implies amyloid tracers of different structures detect different sites on amyloid-ß fibrils or conformations.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Fármacos do Sistema Nervoso Central/farmacocinética , Compostos Radiofarmacêuticos/farmacocinética , Adulto , Idoso , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/patologia , Ligação Competitiva , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Fármacos do Sistema Nervoso Central/química , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Ligação Proteica , Compostos Radiofarmacêuticos/química
12.
Eur J Nucl Med Mol Imaging ; 43(12): 2255-2262, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27514528

RESUMO

This guideline summarizes the current view of the European Association of Nuclear Medicine Drug Development Committee. The purpose of this guideline is to guarantee a high standard of PET studies that are aimed at measuring target occupancy in the brain within the framework of development programs of drugs that act within the central nervous system (CNS drugs). This guideline is intended to present information specifically adapted to European practice. The information provided should be applied within the context of local conditions and regulations.


Assuntos
Encéfalo/metabolismo , Fármacos do Sistema Nervoso Central/farmacocinética , Imagem Molecular/normas , Proteínas do Tecido Nervoso/metabolismo , Tomografia por Emissão de Pósitrons/normas , Guias de Prática Clínica como Assunto , Fármacos do Sistema Nervoso Central/administração & dosagem , Desenho de Fármacos , Monitoramento de Medicamentos/normas , União Europeia
13.
Mol Pharm ; 13(7): 2492-6, 2016 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-27218460

RESUMO

sec-Butylpropylacetamide (SPD) is the amide derivative of valproic acid (VPA). SPD possess a wide-spectrum anticonvulsant profile better than that of VPA and blocks status epilepticus (SE) induced by pilocarpine and organophosphates. The activity of SPD on SE is better than that of benzodiazepines (BZDs) in terms of the ability to block SE when given 20-60 min after the beginning of a seizure. However, intraperitoneal (i.p.) administration to rats cannot be extrapolated to humans. Consequently, in the current study a comparative pharmacokinetic (PK)-pharmacodynamic analysis of SPD was conducted following i.p., intramuscular (i.m.), and intravenous (i.v.) administrations to rats. SPD brain and plasma levels were quantified at various times after dosing following i.p. (60 mg/kg), i.v. (60 mg/kg), and i.m. administrations (120 mg/kg) to rats, and the major PK parameters of SPD were estimated. The antiseizure (SE) efficacies of SPD and its individual stereoisomers were assessed in the pilocarpine-induced BZD-resistant SE model following i.p. and i.m. administrations to rats at 30 min after seizure onset. The absolute bioavailabilities of SPD following i.p. and i.m. administrations were 76% (i.p.) and 96% (i.p.), and its clearance and half-life were 1.8-1.5 L h(-1) kg(-1) and 0.5-1.7 h, respectively. The SPD brain-to-plasma AUC ratios were 1.86 (i.v.), 2.31 (i.p.), and 0.77 (i.m.). Nevertheless, the ED50 values of SPD and its individual stereoisomers were almost identical in the rat pilocarpine-induced SE model following i.p. and i.m. administrations. In conclusion, in rats SPD is completely or almost completely absorbed after i.m. and i.p. administration and readily penetrates into the brain. Consequently, in spite of PK differences, the activities of SPD in the BZD-resistant SE model following i.m. and i.p. administrations are similar.


Assuntos
Amidas/farmacocinética , Anticonvulsivantes/farmacocinética , Encéfalo/metabolismo , Fármacos do Sistema Nervoso Central/farmacocinética , Estado Epiléptico/tratamento farmacológico , Estado Epiléptico/metabolismo , Ácido Valproico/análogos & derivados , Administração Intravenosa , Amidas/administração & dosagem , Amidas/uso terapêutico , Animais , Anticonvulsivantes/administração & dosagem , Anticonvulsivantes/uso terapêutico , Benzodiazepinas/administração & dosagem , Benzodiazepinas/farmacocinética , Benzodiazepinas/uso terapêutico , Encéfalo/efeitos dos fármacos , Fármacos do Sistema Nervoso Central/administração & dosagem , Fármacos do Sistema Nervoso Central/uso terapêutico , Cromatografia Gasosa-Espectrometria de Massas , Injeções Intraperitoneais , Ratos , Ácido Valproico/administração & dosagem , Ácido Valproico/farmacocinética , Ácido Valproico/uso terapêutico
14.
Bioorg Med Chem Lett ; 26(2): 257-261, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26707399

RESUMO

The synthesis, SAR, and preclinical characterization of a series of substituted 6,7-dihydro[1,2,4]triazolo[4,3]pyrazin-8(5H)-one P2X7 receptor antagonists are described. Optimized leads from this series comprise some of the most potent human P2X7R antagonists reported to date (IC50s<1nM). They also exhibit sufficient potency and oral bioavailability in rat to enable extensive in vivo profiling. Although many of the disclosed compounds are peripherally restricted, compound 11d is brain penetrant and upon oral administration demonstrated dose-dependent target engagement in rat hippocampus as determined by ex vivo receptor occupancy with radiotracer 5 (ED50=0.8mg/kg).


Assuntos
Fármacos do Sistema Nervoso Central/farmacologia , Antagonistas do Receptor Purinérgico P2X/farmacologia , Pirazinas/farmacologia , Triazóis/farmacologia , Animais , Barreira Hematoencefálica/metabolismo , Células CACO-2 , Fármacos do Sistema Nervoso Central/síntese química , Fármacos do Sistema Nervoso Central/farmacocinética , Hipocampo/metabolismo , Humanos , Microssomos Hepáticos/metabolismo , Antagonistas do Receptor Purinérgico P2X/síntese química , Antagonistas do Receptor Purinérgico P2X/farmacocinética , Pirazinas/síntese química , Pirazinas/farmacocinética , Ratos , Receptores Purinérgicos P2X7/metabolismo , Estereoisomerismo , Relação Estrutura-Atividade , Triazóis/síntese química , Triazóis/farmacocinética , Trítio
15.
Drug Discov Today Technol ; 20: 27-34, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27986220

RESUMO

To systematically study the pharmacodynamics of a CNS drug early in the development process, we developed and validated a battery of drug-sensitive CNS tests, which we call NeuroCart. Using this test battery, data-intensive phase 1 studies in healthy subjects can be performed to demonstrate the specific, time- and dose-dependent, neurophysiological and/or neuropsychological effects of a compound, thereby confirming whether the test compound reaches its intended target in the CNS - or does not reach its intended target. We use this test battery to demonstrate that a compound passes the blood-brain barrier.


Assuntos
Barreira Hematoencefálica/metabolismo , Testes Neuropsicológicos , Transporte Biológico , Fármacos do Sistema Nervoso Central/farmacocinética , Fármacos do Sistema Nervoso Central/farmacologia , Descoberta de Drogas , Humanos
16.
Int J Neuropsychopharmacol ; 18(10): pyv036, 2015 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-25813017

RESUMO

BACKGROUND: Positron emission tomography microdosing of radiolabeled drugs allows for noninvasive studies of organ exposure in vivo. The aim of the present study was to examine and compare the brain exposure of 12 commercially available CNS drugs and one non-CNS drug. METHODS: The drugs were radiolabeled with (11)C (t 1/2 = 20.4 minutes) and examined using a high resolution research tomograph. In cynomolgus monkeys, each drug was examined twice. In rhesus monkeys, a first positron emission tomography microdosing measurement was repeated after preadministration with unlabeled drug to examine potential dose-dependent effects on brain exposure. Partition coefficients between brain and plasma (KP) were calculated by dividing the AUC0-90 min for brain with that for plasma or by a compartmental analysis (VT). Unbound KP (KP u,u) was obtained by correction for the free fraction in brain and plasma. RESULTS: After intravenous injection, the maximum radioactivity concentration (C max, %ID) in brain ranged from 0.01% to 6.2%. For 10 of the 12 CNS drugs, C max, %ID was >2%, indicating a preferential distribution to brain. A lower C max, %ID was observed for morphine, sulpiride, and verapamil. K P ranged from 0.002 (sulpiride) to 68 (sertraline) and 7 of 13 drugs had KP u,u close to unity. For morphine, sulpiride, and verapamil, K P u,u was <0.3, indicating impaired diffusion and/or active efflux. Brain exposure at microdosing agreed with pharmacological dosing conditions for the investigated drugs. CONCLUSIONS: This study represents the largest positron emission tomography study on brain exposure of commercially available CNS drugs in nonhuman primates and may guide interpretation of positron emission tomography microdosing data for novel drug candidates.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/diagnóstico por imagem , Fármacos do Sistema Nervoso Central/farmacocinética , Morfina/farmacocinética , Sulpirida/farmacocinética , Verapamil/farmacocinética , Animais , Encéfalo/metabolismo , Radioisótopos de Carbono , Fármacos do Sistema Nervoso Central/administração & dosagem , Relação Dose-Resposta a Droga , Feminino , Macaca fascicularis , Macaca mulatta , Modelos Biológicos , Modelos Químicos , Morfina/administração & dosagem , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Sulpirida/administração & dosagem , Verapamil/administração & dosagem
17.
Mol Pharm ; 12(2): 520-32, 2015 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-25496026

RESUMO

In the present work we sought to gain a mechanistic understanding of the physicochemical properties that influence the transport of unbound drug across the blood-brain barrier (BBB) as well as the intra- and extracellular drug exposure in the brain. Interpretable molecular descriptors that significantly contribute to the three key neuropharmacokinetic properties related to BBB drug transport (Kp,uu,brain), intracellular accumulation (Kp,uu,cell), and binding and distribution in the brain (Vu,brain) for a set of 40 compounds were identified using partial least-squares (PLS) analysis. The tailoring of drug properties for improved brain exposure includes decreasing the polarity and/or hydrogen bonding capacity. The design of CNS drug candidates with intracellular targets may benefit from an increase in basicity and/or the number of hydrogen bond donors. Applying this knowledge in drug discovery chemistry programs will allow designing compounds with more desirable CNS pharmacokinetic properties.


Assuntos
Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Fármacos do Sistema Nervoso Central/metabolismo , Fármacos do Sistema Nervoso Central/farmacocinética , Animais , Humanos , Masculino , Modelos Estatísticos , Ratos , Ratos Sprague-Dawley
18.
Pharm Res ; 32(4): 1161-85, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25446769

RESUMO

The blood-brain barrier is a unique cell-based restrictive barrier that prevents the entry of many substances, including most therapeutics, into the central nervous system. A wide range of nanoparticulate delivery systems have been investigated with the aim of targeting therapeutics (drugs, nucleic acids, proteins) to the brain following administration by various routes. This review provides a comprehensive description of the design and formulation of these nanoparticles including the rationale behind individual approaches. In addition, the ability of currently available in-vitro BBB models to accurately predict the in-vivo performance of targeted nanoparticles is critically assessed.


Assuntos
Barreira Hematoencefálica/metabolismo , Fármacos do Sistema Nervoso Central/administração & dosagem , Fármacos do Sistema Nervoso Central/farmacocinética , Portadores de Fármacos/química , Endocitose/fisiologia , Nanopartículas/química , Animais , Transporte Biológico , Células Cultivadas , Humanos , Modelos Biológicos
19.
Eur J Clin Pharmacol ; 71(11): 1333-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26350521

RESUMO

PURPOSE: This study aimed to determine the effect of PET scan timings on the reliability of occupancy parameter estimates and to identify the scan timing design that gives the most reliable occupancy parameter estimates. METHODS: We compared the performance of designs with various sets of sampling time points using the stochastic simulation and estimation method in Perl-speaks-NONMEM. Biases, relative standard errors, relative estimation errors, and root mean square errors were used to compare the performance of designs. RESULTS: Unlike the results of a previous report, we found that rather complicated designs where each subject or group of subjects are allocated to different scan timings were not superior to the simple, conventional fixed-time designs regardless of whether effect compartment or receptor binding models were used. CONCLUSIONS: We conclude that the conventional fixed-time designs that have been used so far may give robust PD parameter estimates for occupancy data obtained from human PET studies of CNS drugs.


Assuntos
Fármacos do Sistema Nervoso Central/farmacocinética , Tomografia por Emissão de Pósitrons/métodos , Receptores de Droga/metabolismo , Fármacos do Sistema Nervoso Central/sangue , Simulação por Computador , Humanos , Modelos Biológicos , Fatores de Tempo
20.
Angew Chem Int Ed Engl ; 54(13): 3967-72, 2015 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-25650865

RESUMO

The blood-brain barrier (BBB) is a formidable physical and enzymatic barrier that tightly controls the passage of molecules from the blood to the brain. In fact, less than 2 % of all potential neurotherapeutics are able to cross it. Here, by applying the retro-enantio approach to a peptide that targets the transferrin receptor, a full protease-resistant peptide with the capacity to act as a BBB shuttle was obtained and thus enabled the transport of a variety of cargos into the central nervous system.


Assuntos
Barreira Hematoencefálica/metabolismo , Peptídeos/síntese química , Peptídeos/farmacocinética , Animais , Transporte Biológico , Bovinos , Fármacos do Sistema Nervoso Central/farmacocinética , Técnicas de Cocultura , Células Endoteliais/metabolismo , Camundongos , Peptídeo Hidrolases/química , Permeabilidade , Ratos , Receptores da Transferrina/efeitos dos fármacos , Estereoisomerismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA