Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Sci Rep ; 8(1): 14421, 2018 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-30258062

RESUMO

DNA damaging therapies can spur the formation of therapy-related cancers, due to mis-repair of lesions they create in non-cancerous cells. This risk may be amplified in patients with impaired DNA damage responses. We disabled key DNA damage response pathways using genetic and pharmacological approaches, and assessed the impact of these deficiencies on the mutagenicity of chemotherapy drugs or the "Smac mimetic" GDC-0152, which kills tumor cells by targeting XIAP, cIAP1 and 2. Doxorubicin and cisplatin provoked mutations in more surviving cells deficient in ATM, p53 or the homologous recombination effector RAD51 than in wild type cells, but suppressing non-homologous end joining (NHEJ) by disabling DNA-PKcs prevented chemotherapy-induced mutagenesis. Vincristine-induced mutagenesis required p53 and DNA-PKcs but was not affected by ATM status, consistent with it provoking ATM-independent p53-mediated activation of caspases and CAD, which creates DNA lesions in surviving cells that could be mis-repaired by NHEJ. Encouragingly, GDC-0152 failed to stimulate mutations in cells with proficient or defective DNA damage response pathways. This study highlights the elevated oncogenic risk associated with treating DNA repair-deficient patients with genotoxic anti-cancer therapies, and suggests a potential advantage for Smac mimetic drugs over traditional therapies: a reduced risk of therapy-related cancers.


Assuntos
Materiais Biomiméticos/farmacologia , Cicloexanos/farmacologia , Dano ao DNA , Sistemas de Liberação de Medicamentos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Mitocondriais , Proteínas de Neoplasias , Neoplasias , Pirróis/farmacologia , Proteínas Reguladoras de Apoptose , Linhagem Celular Tumoral , Reparo do DNA por Junção de Extremidades/efeitos dos fármacos , Células HEK293 , Humanos , Mutagênese/efeitos dos fármacos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia
2.
J Econ Entomol ; 109(6): 2424-2427, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27694506

RESUMO

Current recommendations for applying the antiaggregation pheromone 3-methylcyclohex-2-en-1-one (MCH) to protect live trees from Douglas-fir beetle, Dendroctonus pseudotsugae Hopkins, infestation are to space individual passive releasers (MCH bubble capsules) on a 12- by 12-m grid throughout areas to be protected. Previous field studies and a theoretical study using a puff dispersion model to predict pheromone concentrations have shown that releasers emitting higher rates of MCH spaced farther apart may be as effective as the established standard treatment. During 2012 and 2013, we tested higher release rates of MCH at correspondingly wider spacings to keep the total amount of MCH released per unit area equal in all treatments. In 2012 near Challis, ID, treatments included the established standard release rate and spacing, four and six times the standard release rate at correspondingly wider spacings, and an untreated control. In 2013 near Ketchum, ID, treatments included the established standard release rate and spacing, five and seven times the standard release rate at correspondingly wider spacings, and an untreated control. Results from both years indicated that all MCH treatments were equally effective in reducing Douglas-fir beetle infestation. Using higher release rate formulations at wider spacings will reduce labor costs of installing MCH treatments, and, in cases where it is necessary, retrieving the releasers as well. In addition to reducing labor costs, the revised treatment protocol may increase the feasibility of treating areas that currently may not be possible due to treatment costs.


Assuntos
Cicloexanos/farmacologia , Controle de Insetos/métodos , Feromônios/farmacologia , Pseudotsuga , Gorgulhos/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Agricultura Florestal/economia , Agricultura Florestal/métodos , Controle de Insetos/economia , Pseudotsuga/crescimento & desenvolvimento
3.
J Gene Med ; 14(7): 491-500, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22711445

RESUMO

BACKGROUND: The nuclear membrane of differentiated airway epithelial cells is a significant barrier for nonviral vectors. Trans-cyclohexane-1,2-diol (TCHD) is an amphipathic alcohol that has been shown to collapse nuclear pore cores and allow the uptake of macromolecules that would otherwise be too large for nuclear entry. Previous studies have shown that TCHD can increase lipid-mediated transfection in vitro. METHODS: We aimed to reproduce these in vitro studies using the cationic lipid GL67A, which we are currently assessing in cystic fibrosis trials and, more importantly, we assessed the effects of TCHD on transfection efficiency in differentiated airway epithelium ex vivo and in mouse lung in vivo using three different drug delivery protocols (nebulisation and bolus administration of TCHD to the mouse lung, as well as perfusion of TCHD to the nasal epithelium, which prolongs contact time between the airway epithelium and drug). RESULTS: TCHD (0.5-2%) dose-dependently increased Lipofectamine 2000 and GL67A-mediated transfection of 293T cells by up to 2 logs. Encouragingly, exposure to 8% TCHD (but not 0.5% or 2.0%) increased gene expression in fully differentiated human air liquid interface cultures by approximately 20-fold, although this was accompanied by significant cell damage. However, none of the TCHD treated mice in any of the three protocols had higher gene expression compared to no TCHD controls. CONCLUSIONS: Although TCHD significantly increases gene transfer in cell lines and differentiated airway epithelium ex vivo, this effect is lost in vivo and further highlights that promising in vitro findings often cannot be translated into in vivo applications.


Assuntos
Cicloexanos/farmacologia , Cicloexanóis/farmacologia , Técnicas de Transferência de Genes , Poro Nuclear/efeitos dos fármacos , Sistema Respiratório/efeitos dos fármacos , Animais , Células Cultivadas , Cicloexanos/administração & dosagem , Relação Dose-Resposta a Droga , Vias de Administração de Medicamentos , Epitélio/efeitos dos fármacos , Feminino , Terapia Genética , Vetores Genéticos , Humanos , Lipídeos/farmacologia , Pulmão/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Mucosa Nasal/efeitos dos fármacos , Transfecção
4.
Clin Ther ; 30(7): 1228-50, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18691983

RESUMO

BACKGROUND: The emergence of viral resistance is one of the greatest challenges in the treatment of HIV infection. Maraviroc is the first member of a new class of antiretroviral medications, the CCR5-receptor antagonists. It is approved by the US Food and Drug Administration (FDA) for use in combination with other antiretroviral agents in treatment-experienced patients infected with multidrug-resistant, CCR5-tropic HIV-1. OBJECTIVE: This article provides an overview of the pharmacology, efficacy, and tolerability of maraviroc in the treatment of HIV-1 infection. METHODS: Relevant information was identified through a search of MEDLINE (January 2000-May 2008) using the terms maraviroc, UK-427,857, and CCR5-receptor antagonist. Also consulted were abstracts from the International AIDS Society Conference, the Conference on Retroviruses and Opportunistic Infections, and other relevant scientific meetings. Additional publications were found by searching the reference lists of the identified articles and the FDA Web site. RESULTS: Maraviroc is a selective, reversible, small-molecule CCR5-receptor antagonist. In vitro, it has potent anti-HIV-1 activity, with a mean 90% inhibitory concentration of 2.0 nmol/L. It is widely distributed, with a V(d) of approximately 194 L. Maraviroc is moderately metabolized in the liver (65.3%), primarily via the cytochrome P450 3A4 isozyme. It has an elimination t(1/2) of 15.9 to 22.9 hours. Until more data are available, maraviroc should be avoided in patients with severe hepatic insufficiency; dose adjustment does not appear to be necessary on the basis of age, sex, or renal function. In 2 Phase IIb/III studies, maraviroc 300 mg PO QD or BID was found to be more efficacious than placebo in reducing the viral load at 48 weeks in treatment-experienced, CCR5-tropic HIV-1-infected patients receiving an optimized background regimen (difference vs placebo-QD arm: -0.89 log(10) copies/mL [97.5% CI, -1.17 to -0.62]; BID arm: -1.05 log(10) copies/mL [97.5% CI, -1.33 to -0.78]). The proportion of patients with a viral load < 50 copies/mL was 43.2% in the QD arm and 45.5% in the BID arm, compared with 16.7% in the placebo arm (P < 0.001, both treatment arms vs placebo). In treatment-naive patients infected with CCR5-tropic virus only, maraviroc 300 mg PO BID was not noninferior to oral efavirenz 600 mg QD (difference = -4.2%; lower bound of the 1-sided 97.5% CI, -10.9 [predefined statistical cutoff for noninferiority, -10]). Maraviroc was generally well tolerated in clinical trials. The most frequently reported (> or = 5%) adverse events were upper respiratory tract infection (20.0%), cough (12.7%), pyrexia (12.0%), rash (9.6%), musculoskeletal complaints (8.7%), gastrointestinal and abdominal pain (8.2%), dizziness (8.2%), appetite disorders (7.3%), insomnia (7.0%), herpes infection (6.8%), sinusitis (6.3%), joint complaints (6.1%), bronchitis (5.9%), and constipation (5.4%). The recommended dose of maraviroc differs based on concomitant medications, ranging from 150 to 600 mg BID. CONCLUSION: When used in combination with other antiretroviral agents, maraviroc appears to be a promising agent for treatment-experienced patients infected with multidrug-resistant, CCR5-tropic HIV-1.


Assuntos
Fármacos Anti-HIV , Antagonistas dos Receptores CCR5 , Cicloexanos , Infecções por HIV/tratamento farmacológico , HIV-1 , Triazóis , Fármacos Anti-HIV/economia , Fármacos Anti-HIV/farmacocinética , Fármacos Anti-HIV/farmacologia , Fármacos Anti-HIV/uso terapêutico , Cicloexanos/economia , Cicloexanos/farmacocinética , Cicloexanos/farmacologia , Cicloexanos/uso terapêutico , Interações Medicamentosas , Farmacorresistência Viral Múltipla , HIV-1/efeitos dos fármacos , Humanos , Maraviroc , Triazóis/economia , Triazóis/farmacocinética , Triazóis/farmacologia , Triazóis/uso terapêutico
5.
J Mol Endocrinol ; 38(4): 455-65, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17446235

RESUMO

The systemic treatment with angiogenesis inhibitor has been shown to result in weight reduction and adipose tissue loss in various models of obesity. To verify the mechanism of CKD-732 (TNP-470 analog) against obesity, we evaluated CKD-732's peripheral and central anti-obesity effects. CKD-732 was injected subcutaneously (s.c.) for 7 days in various animal models and intracerebroventricularly (i.c.v.) in arcuate nucleus (ARC) lesion mice, ob/ob mice, and normal littermates. Modulation of the hypothalamic neuropeptide mRNAs after i.c.v. injection was evaluated in ARC lesion mice and normal littermates. A conditioned taste aversion (CTA) was performed using lithium chloride (LiCl) as a positive control agent in Long-Evans Tokushima Otsuka and Otsuka Long-Evans Tokushima fatty (OLETF) rats. As a result, 7 days of CKD-732 s.c. injection reduced the cumulative food intake and the body weight significantly in both treated obese (e.g. 114.8 +/- 13.4 g vs 170.7 +/- 20.6 g, 7.9 +/- 0.5% decrease vs 0.3 +/- 2.2% decrease; in treated OLETF rat versus control OLETF rat, P < 0.01 respectively) and non-obese models. Epididymal and mesenteric fat pads, and the size of adipocytes were significantly decreased in treated rats. A single i.c.v. injection decreased food intake and body weight in ARC lesion mice and ob/ob mice but not in normal littermates. Unexpectedly, the hypothalamic neuropeptide mRNAs were not altered by single i.c.v. injection. CKD-732 also induced a dose-dependent CTA comparable with LiCl injection, which is a commonly used agent to produce a CTA. In conclusion, CKD-732 causes significant body weight and appetite reduction, possibly by decreasing adiposity directly and inducing central anorexia, which is partly explained by a CTA. These results should be carefully verified to assess the utility of CKD-732 as an anti-obesity drug.


Assuntos
Fármacos Antiobesidade/farmacologia , Cinamatos/farmacologia , Cicloexanos/farmacologia , Compostos de Epóxi/farmacologia , Obesidade/tratamento farmacológico , Sesquiterpenos/farmacologia , Adipócitos/efeitos dos fármacos , Adipócitos/patologia , Tecido Adiposo/efeitos dos fármacos , Animais , Fármacos Antiobesidade/administração & dosagem , Fármacos Antiobesidade/uso terapêutico , Núcleo Arqueado do Hipotálamo/patologia , Peso Corporal/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Cinamatos/administração & dosagem , Cinamatos/uso terapêutico , Cicloexanos/administração & dosagem , Cicloexanos/química , Cicloexanos/uso terapêutico , Ingestão de Alimentos/efeitos dos fármacos , Compostos de Epóxi/administração & dosagem , Compostos de Epóxi/uso terapêutico , Hipotálamo/metabolismo , Cloreto de Lítio/farmacologia , Masculino , Camundongos , Camundongos Obesos , Neuropeptídeos/metabolismo , O-(Cloroacetilcarbamoil)fumagilol , Ratos , Ratos Endogâmicos OLETF , Sesquiterpenos/administração & dosagem , Sesquiterpenos/química , Sesquiterpenos/uso terapêutico , Paladar/efeitos dos fármacos
6.
J Parasitol ; 93(6): 1505-11, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18314700

RESUMO

New and efficient methods to screen antibiotics are needed to counter increased antibiotic resistance in pathogens and the emergence of new diseases. Here we report a new insect model for screening antibiotics in vivo using the grasshopper Romalea microptera. The system is inexpensive, efficient, and flexible, avoids animal-welfare problems, and can be used to test against most major pathogenic groups. We employed this system to test 11 commercial antibiotics against a pathogenic Encephalitozoon species (Microsporidia). Oral treatment with fumagillin or thiabendazole significantly reduced pathogen spore counts, whereas spore counts of grasshoppers fed with albendazole, ampicillin, chloramphenicol, griseofulvin, metronidazole, sulfadimethoxine, or tetracycline were not significantly different from the infected controls. Quinine produced a distinct, but nonsignificant, reduction in spores, and streptomycin a nonsignificant increase in spores. Although 2 antibiotics significantly reduced spore counts, in no case was the pathogen totally eliminated. This study demonstrates the validity of this system as a method to screen antibiotics. It also corroborates the difficulty researchers and physicians have had in treating microsporidia infections, and suggests that quinine and related alkaloid compounds should be further examined as possible therapeutic agents against this group of ubiquitous pathogens. In addition, streptomycin and related compounds should be tested to determine if this widely used antibiotic enhances microsporidiosis.


Assuntos
Anti-Infecciosos/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Encephalitozoon/efeitos dos fármacos , Gafanhotos/microbiologia , Análise de Variância , Animais , Antifúngicos/farmacologia , Custos e Análise de Custo , Cicloexanos/farmacologia , Avaliação Pré-Clínica de Medicamentos/economia , Avaliação Pré-Clínica de Medicamentos/ética , Encephalitozoon/crescimento & desenvolvimento , Ácidos Graxos Insaturados/farmacologia , Masculino , Modelos Animais , Quinina/farmacologia , Sesquiterpenos/farmacologia , Tiabendazol/farmacologia
7.
J Acquir Immune Defic Syndr ; 42(2): 183-91, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16639345

RESUMO

OBJECTIVES: To use a viral dynamics model to compare the effectiveness of in vivo viral inhibition of several doses of maraviroc (MVC;UK-427,857) and to use a modeling approach to support design decisions for a monotherapy study using various dosing regimens of maraviroc given with and without food. DESIGN: The pharmacokinetic-pharmacodynamic model was developed using clinical data from a first monotherapy study (study A4001007). This was a randomized, double-blind, placebo-controlled, multicenter study of maraviroc in 44 asymptomatic HIV-1-infected patients. Patients received maraviroc under food restrictions at 25 mg once daily or 50, 100, or 300 mg twice daily, or placebo for 10 days. METHODS: Antiviral responses were assessed by measuring plasma HIV-1 RNA levels during screening, during randomization, at baseline, and daily during the 10 days of treatment and at days 11 to 15, 19, 22, 25, and 40. An integrated pharmacokinetic-pharmacodynamic model was developed using the mixed effects modeling approach with patients' pharmacokinetic profiles on the last day of treatment, HIV-1 RNA levels over time, and the individual viral susceptibility. The parameters derived from the viral dynamic model were used to calculate average viral inhibition fraction, decay rate of actively infected cells, and basic reproductive ratio for each treatment group. Monte Carlo simulation was then used to determine the distribution of viral load change across simulated patients over time for each regimen to be studied in another monotherapy study, A4001015. RESULTS: The decline rate in the 300 mg twice daily group was comparable to that induced by potent protease inhibitor monotherapy, but was significantly slower than that in patients receiving combination therapy including both protease inhibitor and reverse transcriptase inhibitors. The efficacy of inhibition in vivo was estimated to range from 0.15 to 0.38 for the 25 mg once daily dose group and from 0.88 to 0.96 for the 300 mg twice daily dose group. CONCLUSIONS: The model has aided the analysis and interpretation of the clinical data. The use of a model-based approach for selecting doses can accelerate drug development by replacing some arms or trials with simulations.


Assuntos
Fármacos Anti-HIV/administração & dosagem , Fármacos Anti-HIV/farmacocinética , Simulação por Computador , Cicloexanos/administração & dosagem , Cicloexanos/farmacocinética , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Triazóis/administração & dosagem , Triazóis/farmacocinética , Fármacos Anti-HIV/farmacologia , Cicloexanos/farmacologia , Relação Dose-Resposta a Droga , Alimentos , Infecções por HIV/virologia , Humanos , Maraviroc , Método de Monte Carlo , Placebos , RNA Viral/sangue , Triazóis/farmacologia , Carga Viral
8.
Value Health ; 7(1): 13-21, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14720127

RESUMO

OBJECTIVE: The objective of this study was to describe an approach to modeling the efficiency of an intervention by focusing on an established intermediate end point directly. A case study addresses the economic efficiency of obtaining dual glycemic control over time, according to initial choice of treatment. METHODS: From the perspective of a payer in the United States, instead of the usual approach of basing the model on projecting long-term diabetic complications from glycemic control, this model focuses directly on glycemic control. Treatment changes and associated health-care utilization needed to address postprandial glucose. After assigning each of 10000 drug-naïve patients, HbA1c, age, race, and sex based on distributions from a randomized clinical trial, the model applies the efficacy of nateglinide compared to metformin. Sensitivity analyses were carried out for all parameters. Costs are reported in year 2000 US dollars and discounted at 3%. RESULTS: In the base case, starting on nateglinide and increasing the time in dual glycemic control over 3 years by 2.4 months led to savings of US dollars 295 compared to starting on metformin. Savings increased with stricter treatment criteria but decreased if glycemic control was better initially. CONCLUSIONS: This study illustrates the use of an efficiency model that focuses directly on the relevant short-term end point: glycemic control. Starting patients with nateglinide is shown to be an efficient way of obtaining dual glycemic control during the first 3 years of treatment.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/economia , Hipoglicemiantes/economia , Avaliação de Resultados em Cuidados de Saúde/economia , Fenilalanina/análogos & derivados , Adulto , Idoso , Glicemia/efeitos dos fármacos , Análise Custo-Benefício/métodos , Cicloexanos/economia , Cicloexanos/farmacologia , Diabetes Mellitus Tipo 2/sangue , Quimioterapia Combinada , Feminino , Hemoglobinas Glicadas/metabolismo , Custos de Cuidados de Saúde , Humanos , Hipoglicemiantes/farmacologia , Masculino , Metformina/economia , Metformina/farmacologia , Pessoa de Meia-Idade , Modelos Econométricos , Nateglinida , Estudos de Casos Organizacionais , Fenilalanina/economia , Fenilalanina/farmacologia , Estados Unidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA