Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Mov Disord ; 39(3): 618-622, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38291616

RESUMO

BACKGROUND: Acting on the main target of dopaminergic cells, the striatal γ-aminobutyric acid (GABA)-ergic cells, might be a new way to treat persons with Parkinson's disease (PD). OBJECTIVE: The objective of this study was to assess the efficacy of bumetanide, an Na-K-Cl cotransporter (NKCC1) inhibitor, to improve motor symptoms in PD. METHODS: This was a 4-month double-blind, randomized, parallel-group, placebo-controlled trial of 1.75 to 3 mg/day bumetanide as an adjunct to levodopa in 44 participants with PD and motor fluctuations. RESULTS: Compared to the baseline, the mean change in OFF Movement Disorder Society Unified Parkinson's Disease Rating Scale Part III score after 4 months of treatment (primary endpoint) did not improve significantly compared with placebo. No changes between participants treated with bumetanide and those treated with placebo were observed for most other outcome measures. Despite no relevant safety signals, bumetanide was poorly tolerated. CONCLUSIONS: There was no evidence in this study that bumetanide has efficacy in improving motor symptoms of PD. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Assuntos
Doença de Parkinson , Humanos , Doença de Parkinson/tratamento farmacológico , Antiparkinsonianos , Bumetanida/uso terapêutico , Levodopa/uso terapêutico , Avaliação de Resultados em Cuidados de Saúde , Método Duplo-Cego , Resultado do Tratamento
2.
Child Psychiatry Hum Dev ; 54(5): 1360-1372, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-35292925

RESUMO

The efficacy of bumetanide (oral liquid formulation 0.5 mg bid) as a treatment for the core symptoms of autism spectrum disorders in children and adolescents aged 7-17 years is being investigated in an international, randomised, double-blind, placebo-controlled phase III study. The primary endpoint is the change in Childhood Autism Rating Scale 2 (CARS2) total raw score after 6 months of treatment. At baseline, the 211 participants analysed are broadly representative of autistic subjects in this age range: mean (SD) age, 10.4 (3.0) years; 82.5% male; 47.7% with intelligence quotient ≥ 70. Mean CARS2 score was 40.1 (4.9) and mean Social Responsiveness Scale score was 116.7 (23.4). Final study results will provide data on efficacy and safety of bumetanide in autistic children and adolescents.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Humanos , Criança , Masculino , Adolescente , Feminino , Transtorno do Espectro Autista/tratamento farmacológico , Bumetanida/efeitos adversos , Transtorno Autístico/diagnóstico , Método Duplo-Cego , Resultado do Tratamento
3.
Autism Res ; 16(10): 2021-2034, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37794745

RESUMO

The efficacy and safety of bumetanide oral solution for the treatment of autism spectrum disorder (ASD) in children and adolescents was evaluated in two international, multi-center, randomized, double-blind, placebo-controlled phase III trials; one enrolled patients aged 7-17 years (SIGN 1 trial) and the other enrolled younger patients aged 2-6 years (SIGN 2). In both studies, patients were randomized to receive bumetanide oral solution twice daily (BID) or placebo BID during a 6-month double-blind treatment period. The primary endpoint was change in Childhood Autism Rating Scale 2 (CARS2) total raw score from baseline to Week 26. Key secondary endpoints included changes in Social Responsiveness Scale-2, Clinical Global Impression Scale, and Vineland Adaptive Behavior Scale. Each study enrolled 211 patients (bumetanide, n = 107; placebo, n = 104). Both studies were terminated early due to absence of any significant difference between bumetanide and placebo in the overall studied populations. In both studies, CARS2 total raw score decreased from baseline to Week 26 in the bumetanide and placebo groups, with no statistically significant difference between groups. No differences were observed between treatment groups for any of the secondary efficacy endpoints in either study. In both studies, treatment-emergent adverse events that occurred more frequently with bumetanide than placebo included thirst, polyuria, hypokalemia, and dry mouth. These large phase III trials failed to demonstrate a benefit of bumetanide for the treatment of pediatric ASD compared with placebo. Consequently, the sponsor has discontinued the development of bumetanide for the treatment of this condition. Trial registration: https://clinicaltrials.gov: SIGN 1: NCT03715166; SIGN 2: NCT03715153.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Humanos , Criança , Adolescente , Bumetanida/efeitos adversos , Transtorno do Espectro Autista/tratamento farmacológico , Resultado do Tratamento , Método Duplo-Cego
4.
J Autism Dev Disord ; 51(8): 2959-2972, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33151500

RESUMO

There are currently no approved pharmacological treatments to improve social reciprocity and limit repetitive and rigid behaviors in autism spectrum disorder (ASD). We describe the design of two Phase III studies evaluating the efficacy/safety of bumetanide oral liquid formulation in ASD. These are international, multicenter, randomized, double-blind, placebo-controlled studies in children and adolescents with ASD aged 7 to 17 years (n = 200; study 1), or younger children with ASD aged 2 to 6 years (n = 200; study 2). The primary endpoint of each is change in Childhood Autism Rating Scale 2 total raw score after 6 months. These studies could contribute to the first pharmacological treatment to improve social reciprocity and limit repetitive and rigid behaviors in children and adolescents with ASD.


Assuntos
Transtorno do Espectro Autista/tratamento farmacológico , Bumetanida/uso terapêutico , Projetos de Pesquisa , Comportamento Social , Adolescente , Bumetanida/administração & dosagem , Criança , Pré-Escolar , Método Duplo-Cego , Humanos , Masculino , Resultado do Tratamento
5.
Clin Cancer Res ; 14(4): 1258-65, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18281561

RESUMO

PURPOSE: There is a clear clinical need for cytotoxic drugs with a lower systemic toxicity. DTS-201 (CPI-0004Na) is a peptidic prodrug of doxorubicin that shows an improved therapeutic index in experimental models. The purpose of the current study was to complete its preclinical characterization before initiation of phase I clinical trials. EXPERIMENTAL DESIGN: The preclinical development program consisted of a detailed assessment of the general and cardiac toxicity profiles of DTS-201 in mice, rats, and dogs, together with mass balance and antitumoral efficacy studies in rodents. Neprilysin and thimet oligopeptidase expression, two enzymatic activators of DTS-201, was also characterized in human breast and prostate tumor biopsies. RESULTS: The target organs of DTS-201 toxicity in rodents and dogs are typically those of doxorubicin, albeit at much higher doses. Importantly, chronic treatment with DTS-201 proved to be significantly less cardiotoxic than with doxorubicin at doses up to 8-fold higher in rats. The mass balance study showed that [14C] DTS-201 does not accumulate in the body after intravenous administration. The improved therapeutic index of DTS-201 compared with free doxorubicin was confirmed in three tumor xenograft models of prostate, breast, and lung cancer. Neprilysin and/or thimet oligopeptidase are expressed in all experimental human tumor types thus far tested as well as in a large majority of human breast and prostate tumor biopsies. CONCLUSION: DTS-201 gave promising results in terms of general toxicity, cardiovascular tolerance, and in vivo efficacy in xenograft mouse models compared with free doxorubicin. Taken together, these results and the confirmation of the presence of activating enzymes in human tumor biopsies provide a strong rationale for a phase I clinical study in cancer patients.


Assuntos
Doxorrubicina/análogos & derivados , Doxorrubicina/metabolismo , Neoplasias Experimentais/tratamento farmacológico , Oligopeptídeos/farmacologia , Pró-Fármacos/farmacologia , Animais , Neoplasias da Mama/enzimologia , Linhagem Celular Tumoral , Cães , Doxorrubicina/farmacologia , Feminino , Humanos , Masculino , Metaloendopeptidases/biossíntese , Camundongos , Neprilisina/biossíntese , Neoplasias da Próstata/enzimologia , Ratos , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Clin Cancer Res ; 14(7): 2145-53, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18381956

RESUMO

PURPOSE: Irinotecan is a prodrug converted to the active cytotoxic molecule SN38 predominantly by the action of liver carboxylesterases. The efficacy of irinotecan is limited by this hepatic activation that results in a low conversion rate, high interpatient variability, and dose-limiting gastrointestinal toxicity. The purpose of this study was to evaluate a novel peptidic prodrug of SN38 (DTS-108) developed to bypass this hepatic activation and thus reduce the gastrointestinal toxicity and interpatient variability compared with irinotecan. EXPERIMENTAL DESIGN: SN38 was conjugated to a cationic peptide (Vectocell) via an esterase cleavable linker. The preclinical development plan consisted of toxicity and efficacy evaluation in a number of different models and species. RESULTS: The conjugate (DTS-108) is highly soluble, with a human plasma half-life of 400 minutes in vitro. Studies in the dog showed that DTS-108 liberates significantly higher levels of free SN38 than irinotecan without causing gastrointestinal toxicity. In addition, the ratio of the inactive SN38-glucuronide metabolite compared with the active SN38 metabolite is significantly lower following DTS-108 administration, compared with irinotecan, which is consistent with reduced hepatic metabolism. In vivo efficacy studies showed that DTS-108 has improved activity compared with irinotecan. A significant dose-dependent antitumoral efficacy was observed in all models tested and DTS-108 showed synergistic effects in combination with other clinically relevant therapeutic agents. CONCLUSIONS: DTS-108 is able to deliver significantly higher levels of SN38 than irinotecan, without the associated toxicity of irinotecan, resulting in an increased therapeutic window for DTS-108 in preclinical models. These encouraging data merit further preclinical and clinical investigation.


Assuntos
Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacologia , Camptotecina/análogos & derivados , Portadores de Fármacos , Neoplasias Experimentais/tratamento farmacológico , Peptídeos/química , Peptídeos/síntese química , Peptídeos/farmacologia , Pró-Fármacos/farmacologia , Animais , Antineoplásicos Fitogênicos/síntese química , Camptotecina/síntese química , Camptotecina/química , Camptotecina/metabolismo , Camptotecina/farmacologia , Cátions , Cães , Humanos , Irinotecano , Pró-Fármacos/síntese química , Pró-Fármacos/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Biochem J ; 390(Pt 2): 407-18, 2005 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15859953

RESUMO

Short peptide sequences that are able to transport molecules across the cell membrane have been developed as tools for intracellular delivery of therapeutic molecules. This work describes a novel family of cell-penetrating peptides named Vectocell peptides [also termed DPVs (Diatos peptide vectors)]. These peptides, originating from human heparin binding proteins and/or anti-DNA antibodies, once conjugated to a therapeutic molecule, can deliver the molecule to either the cytoplasm or the nucleus of mammalian cells. Vectocell peptides can drive intracellular delivery of molecules of varying molecular mass, including full-length active immunoglobulins, with efficiency often greater than that of the well-characterized cell-penetrating peptide Tat. The internalization of Vectocell peptides has been demonstrated to occur in both adherent and suspension cell lines as well as in primary cells through an energy-dependent endocytosis process, involving cell-membrane lipid rafts. This endocytosis occurs after binding of the cell-penetrating peptides to extracellular heparan sulphate proteoglycans, except for one particular peptide (DPV1047) that partially originates from an anti-DNA antibody and is internalized in a caveolar independent manner. These new therapeutic tools are currently being developed for intracellular delivery of a number of active molecules and their potentiality for in vivo transduction investigated.


Assuntos
Membrana Celular/metabolismo , Portadores de Fármacos/metabolismo , Peptídeos/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação Microbiológicos/genética , Linhagem Celular , Núcleo Celular/química , Núcleo Celular/metabolismo , Citosol/química , Citosol/metabolismo , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Portadores de Fármacos/toxicidade , Humanos , Integrases/metabolismo , Cinética , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/farmacologia , Peptídeos/toxicidade , Transporte Proteico , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Recombinação Genética/genética , Temperatura , Proteínas Virais/metabolismo
9.
J Innate Immun ; 8(4): 330-49, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26987072

RESUMO

(R)-Roscovitine, a pharmacological inhibitor of kinases, is currently in phase II clinical trial as a drug candidate for the treatment of cancers, Cushing's disease and rheumatoid arthritis. We here review the data that support the investigation of (R)-roscovitine as a potential therapeutic agent for the treatment of cystic fibrosis (CF). (R)-Roscovitine displays four independent properties that may favorably combine against CF: (1) it partially protects F508del-CFTR from proteolytic degradation and favors its trafficking to the plasma membrane; (2) by increasing membrane targeting of the TRPC6 ion channel, it rescues acidification in phagolysosomes of CF alveolar macrophages (which show abnormally high pH) and consequently restores their bactericidal activity; (3) its effects on neutrophils (induction of apoptosis), eosinophils (inhibition of degranulation/induction of apoptosis) and lymphocytes (modification of the Th17/Treg balance in favor of the differentiation of anti-inflammatory lymphocytes and reduced production of various interleukins, notably IL-17A) contribute to the resolution of inflammation and restoration of innate immunity, and (4) roscovitine displays analgesic properties in animal pain models. The fact that (R)-roscovitine has undergone extensive preclinical safety/pharmacology studies, and phase I and II clinical trials in cancer patients, encourages its repurposing as a CF drug candidate.


Assuntos
Imunidade Adaptativa , Analgésicos/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Fibrose Cística/tratamento farmacológico , Imunidade Inata , Dor/tratamento farmacológico , Purinas/uso terapêutico , Animais , Ensaios Clínicos como Assunto , Fibrose Cística/imunologia , Humanos , Imunomodulação , Neoplasias/tratamento farmacológico , Roscovitina
10.
Diabetes ; 51(8): 2363-8, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12145146

RESUMO

The effects of benfluorex and two of its metabolites (S 422-1 and S 1475-1) on fatty acid and glucose metabolic fluxes and specific gene expression were studied in hepatocytes isolated from 24-h fasted rats. Both benfluorex and S 422-1 (0.1 or 1 mmol/l) reduced beta-oxidation rates and ketogenesis, whereas S 1475-1 had no effect. At the same concentration, benfluorex and S 422-1 were more efficient in reducing gluconeogenesis from lactate/pyruvate than S 1475-1. Benfluorex inhibited gluconeogenesis at the level of pyruvate carboxylase (45% fall in acetyl-CoA concentration) and of glyceraldehyde-3-phosphate dehydrogenase (decrease in ATP/ADP and NAD(+)/NADH ratios). Accordingly, neither benfluorex nor S 422-1 inhibited gluconeogenesis from dihydroxyacetone, but both stimulated gluconeogenesis from glycerol. In hepatocytes cultured in the presence of benfluorex or S 422-1 (10 or 100 micromol/l), the expression of genes encoding enzymes of fatty acid oxidation (carnitine palmitoyltransferase [CPT] I), ketogenesis (hydroxymethylglutaryl-CoA synthase), and gluconeogenesis (glucose-6-phosphatase, PEPCK) was decreased, whereas mRNAs encoding glucokinase and pyruvate kinase were increased. By contrast, Glut-2, acyl-CoA synthetase, and CPT II gene expression was not affected by benfluorex or S 422-1. In conclusion, this work suggests that benfluorex mainly via S 422-1 reduces gluconeogenesis by affecting gene expression and metabolic status of hepatocytes.


Assuntos
Ácidos Graxos/metabolismo , Fenfluramina/análogos & derivados , Fenfluramina/farmacologia , Regulação Enzimológica da Expressão Gênica , Glucose/metabolismo , Hepatócitos/metabolismo , Acetilcoenzima A/metabolismo , Animais , Carnitina O-Palmitoiltransferase/genética , Jejum , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glucoquinase/genética , Gluconeogênese/efeitos dos fármacos , Glucose-6-Fosfatase/genética , Hepatócitos/efeitos dos fármacos , Hidroximetilglutaril-CoA Sintase/genética , Lactatos/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Oxirredução , Piruvato Quinase/genética , Piruvatos/metabolismo , RNA Mensageiro/genética , Ratos , Serotoninérgicos/farmacologia , Transcrição Gênica/efeitos dos fármacos
11.
Fundam Clin Pharmacol ; 17(1): 61-9, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12588631

RESUMO

The potency of three sulphonylureas, glibenclamide, glimepiride and gliclazide in antagonizing the vasorelaxant action of openers of adenosine triphosphate (ATP)-regulated K+ channel (KATP) was studied in vivo and in vitro in micro- and macrovessels, respectively. In the hamster cheek pouch, the vasodilatation and the increase in vascular diameter and blood flow induced by diazoxide were markedly reduced by the addition of either glibenclamide or glimepiride (0.8 microm) while they were not affected by gliclazide up to 12 microm. Similarly, in rat and guinea-pig isolated aortic rings, glibenclamide, glimepiride and gliclazide reduced the vasodilator activity of cromakalim. However, the inhibitory effect of gliclazide was considerably less when compared with either glimepiride or glibenclamide. These results suggest that, in contrast to glibenclamide and glimepiride, therapeutically relevant concentrations of gliclazide do not block the vascular effects produced by KATP channel openers in various in vitro and in vivo animal models.


Assuntos
Hipoglicemiantes/farmacologia , Canais de Potássio/efeitos dos fármacos , Compostos de Sulfonilureia/farmacologia , Vasodilatação/efeitos dos fármacos , Trifosfato de Adenosina/fisiologia , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/fisiologia , Cricetinae , Cromakalim/antagonistas & inibidores , Cromakalim/farmacologia , Diazóxido/antagonistas & inibidores , Diazóxido/farmacologia , Interações Medicamentosas , Gliclazida/farmacologia , Glibureto/farmacologia , Cobaias , Técnicas In Vitro , Ativação do Canal Iônico , Masculino , Canais de Potássio/fisiologia , Ratos , Ratos Wistar , Especificidade da Espécie , Vasodilatadores/antagonistas & inibidores , Vasodilatadores/farmacologia
12.
J Diabetes Complications ; 17(2 Suppl): 6-10, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12623162

RESUMO

INTRODUCTION: Sulfonylureas are widely prescribed for the treatment of type 2 diabetes. Their therapeutic efficacy resides in the ability to bind to sulfonylurea receptors (SURs) present on the beta-cell plasma membrane, to close the ATP-regulated potassium (K(ATP)) channel, and thereby to enhance glucose-stimulated insulin secretion. These receptors are also found in a wide variety of extra-pancreatic tissues such as brain, peripheral nerves, heart, and vascular smooth muscle where they contribute to the regulation of the vascular tone. OBJECTIVE: The objective of the present study was to determine the potency of three sulfonylureas, glibenclamide, gliclazide, and glimepiride, in antagonizing the vasorelaxant action of diazoxide, an ATP-regulated K(+) channel (K(ATP)) opener, in vivo, using the hamster cheek pouch preparation and evaluating the changes in mean internal diameter and blood flow of arterioles and venules. MATERIAL AND METHODS: Cheek pouches of anesthetized male hamsters superfused with a HEPES-supported HCO(3)(-)-buffered saline solution were placed under an intravital microscope coupled to a closed-circuit TV system. All substances were applied topically. MEASUREMENTS: Mean arteriolar and venular internal diameters using an image shearing device, red blood cell (RBC) velocity by the dual-slit photometric technique and microvessel volume flow was calculated from diameters and RBC velocities. RESULTS: The numbers are given in order, first diameter and then flow, always for the highest concentration of diazoxide tested, by itself or in combination with a given sulfonylurea: (1) diazoxide, used in doses of 0.01, 1, and 100 microM, elicited a dose-dependent dilation and flow increase in arterioles [increase of 52.1% (P<.01) and 41.2% (P<.01)] and venules [37.9% (P<.05) and 57.6% (P<.01)]; (2) glibenclamide (0.81 microM)+diazoxide 29.3% (P=.172) and 25.0% (P=.064) for arterioles and 8% (P=.654) and 3.7% (P=.769) for venules; (3) gliclazide (12 microM)+diazoxide 51.0% (P<.01) and 46.7% (P<.01) for arterioles and 59.0% (P<.01) and 45.2% (P<.01) for venules; (4) glimepiride (0.82 microM)+diazoxide 22.8% (P=.228) and 12.5% (P=.305) for arterioles and 15.6% (P=.415) and 16.0% (P=.291) for venules. CONCLUSION: These results suggest that, in contrast to glibenclamide and glimepiride, therapeutic concentrations of gliclazide produce no cross-reactivity with smooth muscle cell K(ATP) channels in the microvessels of the hamster cheek pouch. Previous studies have confirmed these results in isolated aortic rings of rats and guinea pigs.


Assuntos
Vasos Sanguíneos/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Canais de Potássio/efeitos dos fármacos , Compostos de Sulfonilureia/farmacologia , Vasodilatação/efeitos dos fármacos , Trifosfato de Adenosina/fisiologia , Análise de Variância , Animais , Cricetinae , Diazóxido/farmacologia , Interações Medicamentosas , Gliclazida/farmacologia , Glibureto/farmacologia , Técnicas In Vitro , Ativação do Canal Iônico , Masculino , Mesocricetus , Canais de Potássio/fisiologia , Vasodilatadores/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA