Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Nanobiotechnology ; 18(1): 89, 2020 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-32527262

RESUMO

BACKGROUND: Farnesol is a sesquiterpene from propolis and citrus fruit that shows promising anti-bacterial activity for caries treatment and prevention, but its hydrophobicity limits the clinical application. We aimed to develop the novel polymeric micelles (PMs) containing a kind of derivative of farnesol and a ligand of pyrophosphate (PPi) that mediated PMs to adhere tightly with the tooth enamel. RESULTS: Farnesal (Far) was derived from farnesol and successfully linked to PEG via an acid-labile hydrazone bond to form PEG-hyd-Far, which was then conjugated to PPi and loaded into PMs to form the aimed novel drug delivery system, PPi-Far-PMs. The in vitro test about the binding of PPi-Far-PMs to hydroxyapatite showed that PPi-Far-PMs could bind rapidly to hydroxyapatite and quickly release Far under the acidic conditions. Results from the mechanical testing and the micro-computed tomography indicated that PPi-Far-PMs could restore the microarchitecture of teeth with caries. Moreover, PPi-Far-PMs diminished the incidence and severity of smooth and sulcal surface caries in rats that were infected with Streptococcus mutans while being fed with a high-sucrose diet. The anti-caries efficacy of free Far can be improved significantly by PPi-Far-PMs through the effective binding of it with tooth enamel via PPi. CONCLUSIONS: This novel drug-delivery system may be useful for the treatment and prevention of dental caries as well as the targeting therapy of anti-bacterial drugs in the oral disease.


Assuntos
Cariostáticos , Cárie Dentária , Durapatita , Farneseno Álcool/análogos & derivados , Micelas , Animais , Cariostáticos/química , Cariostáticos/farmacocinética , Cariostáticos/farmacologia , Cárie Dentária/tratamento farmacológico , Cárie Dentária/metabolismo , Cárie Dentária/patologia , Difosfatos/química , Difosfatos/farmacocinética , Difosfatos/farmacologia , Portadores de Fármacos , Durapatita/química , Durapatita/metabolismo , Farneseno Álcool/química , Farneseno Álcool/farmacocinética , Farneseno Álcool/farmacologia , Interações Hidrofóbicas e Hidrofílicas , Dente Molar/efeitos dos fármacos , Dente Molar/ultraestrutura , Polietilenoglicóis/química , Ratos , Streptococcus mutans/efeitos dos fármacos
2.
Cancer Chemother Pharmacol ; 82(3): 511-519, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29992354

RESUMO

PURPOSE: Patients with RAS-positive tumors respond poorly to chemotherapies and have a few treatment options. Salirasib is an oral RAS inhibitor that competitively blocks the membrane association of RAS proteins. The aim of this phase I multiple-ascending-dose clinical trial was to investigate the safety and pharmacokinetics of Salirasib in Japanese patients with relapsed/refractory solid tumors and to explore its efficacy. METHODS: Salirasib was started at a dose of 100-mg twice-daily and escalated to a maximum of 1000-mg twice-daily from days 1 to 21 of a 28-day regimen. The pharmacokinetics was evaluated on days 1 and 21. Dose-limiting toxicity (DLT) and adverse events (AEs) were monitored throughout the trial. Patients with stable disease or better repeated the dosing regimen. RESULTS: A total of 21 patients received Salirasib. Among 14 patients tested, 4 had KRAS mutations. Cmax and AUCinf were maximal at 800 mg. No maximum tolerable dose was discerned, as no DLT was observed in any dosing group. The most frequently observed AEs were gastrointestinal disturbances, including diarrhea, abdominal pain, and nausea. No AEs led to discontinuation. All patients completed the first regimen and 11 patients repeated the regimen (median: 2 cycles; range: 1-13). Patients with KRAS mutations showed median progression-free survival of 227 days (range: 79-373). CONCLUSION: Salirasib was safe and well tolerated in Japanese patients, and 800-mg twice-daily is recommended for phase II trials. Although the number of participants with KRAS mutations was limited, the remarkably long progression-free period warrants further investigation. CLINICAL TRIAL REGISTRATION: JAPIC Clinical Trials Information; JapicCTI-121751.


Assuntos
Farneseno Álcool/análogos & derivados , Neoplasias/tratamento farmacológico , Salicilatos/administração & dosagem , Proteínas ras/antagonistas & inibidores , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/efeitos adversos , Inibidores Enzimáticos/farmacocinética , Farneseno Álcool/administração & dosagem , Farneseno Álcool/efeitos adversos , Farneseno Álcool/farmacocinética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias/metabolismo , Neoplasias/patologia , Salicilatos/efeitos adversos , Salicilatos/farmacocinética
3.
Antimicrob Agents Chemother ; 59(7): 4190-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25941222

RESUMO

Like normal cellular nucleosides, the nucleoside reverse transcriptase (RT) inhibitor (NRTI) 4'-ethynyl-2-fluoro-2'-deoxyadenosine (EFdA) has a 3'-hydroxyl moiety, and yet EFdA is a highly potent inhibitor of human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) replication with activity against a broad range of clinically important drug-resistant HIV isolates. We evaluated the anti-HIV activity of EFdA in primary human cells and in HIV-infected humanized mice. EFdA exhibited excellent potency against HIVJR-CSF in phytohemagglutinin-stimulated peripheral blood mononuclear cells (PBMCs), with a 50% inhibitory concentration of 0.25 nM and a selectivity index of 184,000; similar antiviral potency was found against 12 different HIV clinical isolates from multiple clades (A, B, C, D, and CRF01_AE). EFdA was readily absorbed after oral dosing (5 mg/kg of body weight) in both mice and the rhesus macaque, with micromolar levels of the maximum concentration of drug in serum (Cmax) attained at 30 min and 90 min, respectively. Trough levels were at or above 90% inhibitory concentration (IC90) levels in the macaque at 24 h, suggesting once-daily dosing. EFdA showed reasonable penetration of the blood-brain barrier in the rhesus macaque, with cerebrospinal fluid levels at approximately 25% of plasma levels 8 h after single oral dosing. Rhesus PBMCs isolated 24 h following a single oral dose of 5 mg/kg EFdA were refractory to SIV infection due to sufficiently high intracellular EFdA-triphosphate levels. The intracellular half-life of EFdA-triphosphate in PBMCs was determined to be >72 h following a single exposure to EFdA. Daily oral administration of EFdA at low dosage levels (1 to 10 mg/kg/day) was highly effective in protecting humanized mice from HIV infection, and 10 mg/kg/day oral EFdA completely suppressed HIV RNA to undetectable levels within 2 weeks of treatment.


Assuntos
Fármacos Anti-HIV/uso terapêutico , Compostos de Diazônio/uso terapêutico , Farneseno Álcool/análogos & derivados , Infecções por HIV/tratamento farmacológico , Inibidores da Transcriptase Reversa/uso terapêutico , Animais , Fármacos Anti-HIV/administração & dosagem , Barreira Hematoencefálica , Compostos de Diazônio/administração & dosagem , Compostos de Diazônio/farmacocinética , Farneseno Álcool/administração & dosagem , Farneseno Álcool/farmacocinética , Farneseno Álcool/uso terapêutico , Citometria de Fluxo , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , Meia-Vida , Humanos , Técnicas In Vitro , Macaca mulatta , Camundongos , Camundongos SCID , Monócitos/efeitos dos fármacos , Monócitos/virologia , RNA Viral/biossíntese , RNA Viral/efeitos dos fármacos , Inibidores da Transcriptase Reversa/administração & dosagem , Inibidores da Transcriptase Reversa/farmacocinética , Vírus da Imunodeficiência Símia , Viremia/tratamento farmacológico , Viremia/virologia
4.
J Biol Chem ; 288(38): 27444-27455, 2013 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-23908355

RESUMO

Mammalian cells can use exogenous isoprenols to generate isoprenoid diphosphate substrates for protein isoprenylation, but the mechanism, efficiency, and biological importance of this process are not known. We developed mass spectrometry-based methods using chemical probes and newly synthesized stable isotope-labeled tracers to quantitate incorporation of exogenously provided farnesol, geranylgeraniol, and unnatural analogs of these isoprenols containing an aniline group into isoprenoid diphosphates and protein isoprenylcysteines by cultured human cancer cell lines. We found that at exogenous isoprenol concentrations >10 µM, this process can generate as much as 50% of the cellular isoprenoid diphosphate pool used for protein isoprenylation. Mutational activation of p53 in MDA-MB-231 breast cancer cells up-regulates the mevalonate pathway to promote tumor invasiveness. p53 silencing or pharmacological inhibition of HMG-CoA reductase in these cells decreases protein isoprenylation from endogenously synthesized isoprenoids but enhances the use of exogenous isoprenols for this purpose, indicating that this latter process is regulated independently of the mevalonate pathway. Our observations suggest unique opportunities for design of cancer cell-directed therapies and may provide insights into mechanisms underlying pleiotropic therapeutic benefits and unwanted side effects of mevalonate pathway inhibition.


Assuntos
Antineoplásicos/farmacologia , Diterpenos/farmacologia , Farneseno Álcool/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Ácido Mevalônico/metabolismo , Neoplasias/tratamento farmacológico , Fosfatos de Poli-Isoprenil/metabolismo , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Diterpenos/farmacocinética , Farneseno Álcool/análogos & derivados , Farneseno Álcool/farmacocinética , Humanos , Hidroximetilglutaril-CoA Redutases/genética , Hidroximetilglutaril-CoA Redutases/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacocinética , Mutação , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Prenilação de Proteína/efeitos dos fármacos , Prenilação de Proteína/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
5.
Invest New Drugs ; 30(6): 2391-9, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22547163

RESUMO

PURPOSE: S-trans,trans-Farnesylthiosalicylic Acid (FTS, salirasib) inhibits Ras-dependent cell growth by dislodging all isoforms of Ras, including mutant Ras, from the plasma membrane. This study evaluated the activity, safety, and toxicity of salirasib in preclinical models and patients with metastatic pancreatic adenocarcinoma (PDA). PATIENTS AND METHODS: In the preclinical study, salirasib was tested, alone and in combination with gemcitabine, in patient derived xenografts (PDX) of PDA. In the clinical study, treatment-naïve patients with advanced, metastatic PDA were treated with a standard dose schedule of gemcitabine and salirasib 200-800 mg orally (PO) twice daily (bid) for 21 days every 28 days. Tissue from preclinical models and patients' biopsies were collected pre-treatment and on Cycle (C) 1, Day (D) 9 to characterize the effect of gemcitabine and salirasib on activated Ras protein levels. Plasma samples for pharmacokinetics were collected for salirasib administered alone and in combination. RESULTS: Salirasib inhibited the growth of 2/14 PDX models of PDA and modulated Ras signaling in these tumors. Nineteen patients were enrolled. No DLTs occurred. Common adverse events included hematologic and gastrointestinal toxicities and fatigue. The median overall survival was 6.2 months and the 1 year survival 37 %. In 2 patients in whom paired tissue biopsies were available, Ras and KRas protein levels were decreased on C1D9. Salirasib exposure was not altered by gemcitabine and did not correlate with PD outcomes. CONCLUSION: The combination of gemcitabine and salirasib appears well-tolerated, with no alteration of salirasib exposure, and exerted clinical and PD activity in PDA.


Assuntos
Adenocarcinoma/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/sangue , Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacocinética , Farneseno Álcool/administração & dosagem , Farneseno Álcool/análogos & derivados , Farneseno Álcool/sangue , Farneseno Álcool/farmacocinética , Feminino , Humanos , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas p21(ras) , Salicilatos/administração & dosagem , Salicilatos/sangue , Salicilatos/farmacocinética , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas ras/metabolismo , Gencitabina
6.
Biochem Biophys Res Commun ; 409(4): 752-7, 2011 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-21621522

RESUMO

ZL11n is a novel furoxan-based nitric oxide (NO)-releasing derivative of farnesylthiosalicylic acid. In this study, we examined the anticancer effects and the potential mechanism of action of ZL11n in vitro and in vivo. It was found that ZL11n exhibited a favorable, selective cytotoxic effect in the HepG2 cell line. The yield of NO in the ZL11n treated HepG2 cells was much higher than in the control group and the normal human liver L-02 cells. Furthermore, the NO concentration was correlated to the degree of cytotoxicity observed. The ZL11n-induced apoptosis was assessed by Annexin V-FITC/propidium iodide flow cytometry assay. ZL11n triggered the mitochondrial/caspase apoptotic pathway by decreasing mitochondrial membrane potential, cytochrome c release from mitochondrial, and reducing the Bcl-2-to-Bax ratio, in addition to activating the caspase cascade. Simultaneously, we found that ZL11n treatment led to an increase in JNK and ERK1/2 phosphorylation. Furthermore, treatment with SP600125 (a JNK inhibitor) and PD98059 (an ERK1/2 inhibitor) prior to ZL11n treatment was found to significantly reverse ZL11n-induced apoptosis. The in vivo findings also revealed that ZL11n significantly reduced tumor volume and weight in the H(22) solid tumor mouse model examined. In short, our findings suggest that ZL11n induced apoptosis through the coordination of the mitochondrial apoptotic pathway (activated by NO) and MAPKs signaling pathway (triggered by JNK or ERK).


Assuntos
Antineoplásicos/farmacologia , Apoptose , Carcinoma Hepatocelular/tratamento farmacológico , Farneseno Álcool/análogos & derivados , Neoplasias Hepáticas/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Oxidiazóis/farmacocinética , Animais , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Carcinoma Hepatocelular/enzimologia , Carcinoma Hepatocelular/patologia , Ativação Enzimática , Farneseno Álcool/química , Farneseno Álcool/farmacocinética , Farneseno Álcool/uso terapêutico , Células Hep G2 , Humanos , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Mitocôndrias/enzimologia , Óxido Nítrico/metabolismo , Oxidiazóis/química , Oxidiazóis/uso terapêutico , Salicilatos/química
7.
Future Oncol ; 6(6): 885-91, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20528225

RESUMO

The Ras family of genes is involved in the cellular regulation of proliferation, differentiation, cell adhesion and apoptosis. The K-ras gene is mutated in over 90% of pancreatic cancer cases. Salirasib (S-trans,trans-farnesylthiosalycilic acid [FTS]) is a synthetic small molecule that acts as a potent Ras inhibitor. It is a farnesylcysteine mimetic that selectively disrupts the association of active RAS proteins with the plasma membrane. Animal studies demonstrated that salirasib inhibited tumor growth, downregulated gene expression in the cell cycle and Ras signaling pathways. In a clinical study of salirasib combined with standard doses of gemcitabine, it was demonstrated that the two drugs have no overlapping pharmacokinetics. The salirasib recommended dose was 600 mg twice daily and the progression-free survival was 4.7 months. Future studies will determine whether salirasib adds to the anti-tumor activity of drugs approved by the US FDA for pancreatic cancer.


Assuntos
Antineoplásicos/uso terapêutico , Farneseno Álcool/análogos & derivados , Neoplasias Pancreáticas/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Salicilatos/uso terapêutico , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Ensaios Clínicos Fase I como Assunto , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacocinética , Intervalo Livre de Doença , Ensaios de Seleção de Medicamentos Antitumorais , Farneseno Álcool/administração & dosagem , Farneseno Álcool/efeitos adversos , Farneseno Álcool/farmacocinética , Farneseno Álcool/uso terapêutico , Farnesiltranstransferase/antagonistas & inibidores , Galectinas/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Neoplasias Pancreáticas/enzimologia , Ligação Proteica/efeitos dos fármacos , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/farmacocinética , Proteínas Proto-Oncogênicas p21(ras)/antagonistas & inibidores , Salicilatos/administração & dosagem , Salicilatos/efeitos adversos , Salicilatos/farmacocinética , Transdução de Sinais/efeitos dos fármacos , Gencitabina
8.
Food Chem Toxicol ; 48(1): 18-23, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19747520

RESUMO

The relationship between in vitro and in vivo skin absorption of lipophilic cosmetic ingredients (dibutyl phthalate (DBP, Log K(ow): 4.45), farnesol (Log K(ow): 5.77) and geraniol (Log K(ow): 3.56) from an oil-in-water emulsion was investigated in the hairless guinea pig. In vivo absorption of DBP, farnesol and geraniol 24h after dermal application was 62.0+/-2.0, 39.8+/-2.5, and 15.1+/-1.8% of the applied dose (%AD), respectively. In vitro absorption was measured at 24 and 72 h by using flow-through diffusion cells (0.64 cm(2)) with a receptor fluid consisting of HHBSS+4% BSA. In vitro studies of DBP, farnesol and geraniol absorption over 24h found 27.1+/-1.9, 43.5+/-3.3 and 45.9+/-3.2%AD in receptor fluid, respectively, and over 72 h found 59.9+/-3.2, 77.5+/-7.1 and 49.0+/-6.3%AD, respectively. We found that the amount of DBP absorbed in vivo after 24h closely agreed with the amount of DBP found in the receptor fluid in vitro after 72 h. In contrast, the amount of topically applied farnesol absorbed in vivo after 24h was similar to the amount of farnesol found in receptor fluid in vitro after 24h. A direct comparison between the in vivo absorption of geraniol and the in vitro absorption at 24 and 72 h was not meaningful due to the rapid evaporation of geraniol from the skin. Our in vitro results suggest that lipophilic chemicals initially form a reservoir in skin, and the material in the reservoir may ultimately diffuse out of the skin into the receptor fluid within 72 h. Our results also demonstrate the utility of in vivo studies for resolving questions about the fate of lipophilic chemicals remaining in skin after in vitro absorption studies.


Assuntos
Dibutilftalato/farmacocinética , Farneseno Álcool/farmacocinética , Absorção Cutânea/fisiologia , Terpenos/farmacocinética , Monoterpenos Acíclicos , Animais , Química Farmacêutica , Dibutilftalato/química , Difusão , Emulsões , Farneseno Álcool/química , Feminino , Cobaias , Técnicas In Vitro , Solubilidade , Terpenos/química
9.
Cancer Chemother Pharmacol ; 65(2): 235-41, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19484470

RESUMO

PURPOSE: This phase I first-in-human trial evaluated salirasib, an S-prenyl derivative of thiosalicylic acid that competitively blocks RAS signaling. METHODS: Patients with advanced cancers received salirasib twice daily for 21 days every 4 weeks. Doses were escalated from 100 to 200, 400, 600, and 800 mg. RESULTS: The most common toxicity was dose-related diarrhea (Grade 1-2, 79% of 24 patients). Other toxicities included abdominal pain, nausea, and vomiting. No Grade 3-4 toxicity was noted. Nineteen (79%) patients had no drug-related toxicity >Grade 1. Dose-limiting toxicity (DLT) was not reached, but all three patients treated with 800 mg experienced Grade 1-2 diarrhea, brogating dose escalation. Six patients were treated at a dose of 600 mg with no DLTs. Seven (29%) patients had stable disease on salirasib for ≥4 months (range 4-23+). The salirasib pharmacokinetic profile was characterized by slow absorption and a rapid elimination phase following oral administration. Salirasib exposure (C(max); day 1 AUC(inf) vs. day 15 AUC(0-12 h)) was similar between days 1 and 15 (P > 0.05). The T(1/2) (mean ± SD) was 3.6 ± 2.2 h on day 1. CONCLUSIONS: Salirasib therapy was well tolerated. The recommended dose for phase II studies is 600 mg twice daily.


Assuntos
Antineoplásicos/uso terapêutico , Farneseno Álcool/análogos & derivados , Neoplasias/tratamento farmacológico , Salicilatos/uso terapêutico , Adulto , Idoso , Antineoplásicos/química , Antineoplásicos/farmacocinética , Farneseno Álcool/química , Farneseno Álcool/farmacocinética , Farneseno Álcool/uso terapêutico , Feminino , Humanos , Linfoma/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Salicilatos/química , Salicilatos/farmacocinética , Estereoisomerismo
10.
J Med Chem ; 52(1): 197-205, 2009 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-19072665

RESUMO

The Ras inhibitor S-trans,trans-farnesylthiosalicylic acid (FTS, Salirasib) interferes with Ras membrane interactions that are crucial for Ras-dependent transformation. It remains unknown whether modifications of the carboxyl group of FTS can affect its activity. Here we show that specific modifications of the FTS carboxyl group by esterification or amidation yield compounds with improved growth inhibitory activity, compared to FTS, as shown in Panc-1 and U87 cells. The most potent compounds were FTS-methoxymethyl ester and FTS-amide. However, selectivity toward active Ras-GTP, as known for FTS, was apparent with the amide derivatives of FTS. FTS-amide exhibited the overall highest efficacy in inhibition of Ras-GTP and cell growth. This new compound significantly inhibited growth of both Panc-1 tumors and U87 brain tumors. Thus amide derivatives of the FTS carboxyl group provide potent cell-growth inhibitors without loss of selectivity toward the active Ras protein and may serve as new candidates in cancer therapy.


Assuntos
Antineoplásicos/síntese química , Antineoplásicos/uso terapêutico , Farneseno Álcool/análogos & derivados , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Salicilatos/síntese química , Salicilatos/uso terapêutico , Amidas/química , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Farneseno Álcool/síntese química , Farneseno Álcool/química , Farneseno Álcool/farmacocinética , Farneseno Álcool/uso terapêutico , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Nus , Estrutura Molecular , Neoplasias/enzimologia , Proteína Oncogênica p21(ras)/metabolismo , Salicilatos/química , Salicilatos/farmacocinética , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Transplant Proc ; 40(4): 967-70, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18555091

RESUMO

The competition between the native and the grafted liver in heterotopic auxiliary liver transplantation (HALT) with portal vein arterialization (PVA) was investigated in a rat model. The experimental groups were: HALT with flow-regulated PVA and 70% resection of a native liver and graft (n = 32; group I) versus 70% liver resection (n = 32; group II). After HALT, the weight of the native liver increased until the sixth postoperative week (431% +/- 55% of the intraoperative weight), whereas, the graft weight was only 76% +/- 31% of the intraoperative weight at this time. In group II, liver weight increased continuously to 529% +/- 30% of the intraoperative weight after 6 weeks. On postoperative day 2, there was significantly increased proliferative hepatocellular activity in all groups. This was highest in the resected livers of group II, followed by the native livers of group I, and the grafts of group I (301 +/- 126 vs 262 +/- 97 vs 216 +/- 31 Ki-67-positive hepatocytes/10 visual fields). However, the differences between the groups were not significant. With regard to hepatocellular apoptosis, the livers were similar among all groups and at all time points, M30-positive hepatocyte counts were

Assuntos
Transplante de Fígado/fisiologia , Fígado/fisiologia , Animais , Compostos de Diazônio/farmacocinética , Farneseno Álcool/análogos & derivados , Farneseno Álcool/farmacocinética , Rejeição de Enxerto/fisiopatologia , Antígeno Ki-67/análise , Testes de Função Hepática , Masculino , Modelos Animais , Tamanho do Órgão , Veia Porta/fisiologia , Ratos , Tecnécio/farmacocinética
12.
Cancer Chemother Pharmacol ; 61(1): 89-96, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17909812

RESUMO

BACKGROUND: S-trans,trans-farnesylthiosalicylic acid (salirasib, FTS) is a synthetic small molecule that acts as a potent Ras inhibitor. Salirasib inhibits specifically both oncogenically activated Ras and growth factor receptor-mediated Ras activation, resulting in the inhibition of Ras-dependent tumor growth. The objectives of this study were to develop a sensitive LC-MS/MS assay for determination of FTS in plasma, to assess the bioavailabilty of FTS after oral administration to mice, and then to examine the efficacy of orally administered FTS for inhibition of tumor growth in a nude mouse model. METHODS: FTS was isolated from mouse plasma by liquid chromatography on a Columbus 5-mum particle size, 50 x 2 mm id column with a methanol/5 mM ammonium acetate (80/20) mobile phase (isocratic elution) at a flow rate of 0.3 ml/min. MS/MS was performed on a PE Sciex API 365 with Turbo Ion Spray as interface and negative ion ionization; parent ion (m/z): 357.2; daughter ion (m/z) 153.2; retention time 2.3 min. For plasma analysis, the amount of analyte in each sample was calculated by comparing response of the analyte in that sample to a nine-point standard curve linear over the range 3-1000 ng/ml. Pharmacokinetic studies were performed in mice following intraperitoneal dosing (20 mk/kg in PBS) or oral dosing (40 mg/kg in either 0.5% aqueous CMC or corn oil). Panc-1 tumor growth in nude mice was determined following daily oral dosing with FTS in 0.5% CMC (40, 60, or 80 mg/kg), or in combination with weekly gemcitabine (30 mg/kg). RESULTS: Salirasib was readily detected in mouse plasma by LC-MS/MS at a detection limit of 3 ng/ml. For each route of administration, t (max) was 1 h and t (1/2) ranged from 1.86 to 2.66 h. Compared to IP administration, the oral bioavailabilty of FTS was 69.5% for oral CMC and 55% for oral corn oil suspensions, while clearance and volume of distribution were higher in both oral preparations. The orally administered salirasib inhibited panc-1 tumor growth in a dose dependent manner (67% reduction in tumor weight at the highest dose, P < 0.002 vs. control, n = 10 mice per group) and at a 40 mg/kg daily dose was synergistic with gemcitabine (83% increase in survival rate, n = 8 mice per group). CONCLUSIONS: Salirasib exhibits good bioavailabilty after oral administration, as determined by a highly sensitive method for quantification in plasma. The orally available Ras inhibitor salirasib inhibited growth in nude mice, and may thus be considered for clinical trials.


Assuntos
Antineoplásicos/administração & dosagem , Inibidores Enzimáticos/administração & dosagem , Farneseno Álcool/análogos & derivados , Neoplasias Pancreáticas/tratamento farmacológico , Salicilatos/administração & dosagem , Proteínas ras/antagonistas & inibidores , Administração Oral , Animais , Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Disponibilidade Biológica , Cromatografia Líquida/métodos , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacocinética , Farneseno Álcool/administração & dosagem , Farneseno Álcool/farmacocinética , Injeções Intraperitoneais , Camundongos , Camundongos Nus , Distribuição Aleatória , Salicilatos/farmacocinética , Solventes/química , Taxa de Sobrevida , Espectrometria de Massas em Tandem/métodos , Gencitabina
13.
Arch Biochem Biophys ; 448(1-2): 93-103, 2006 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-16307722

RESUMO

In a previous study (A. Hemmerlin, T.J. Bach, Plant Physiol. 123 (2000) 1257-1268), we have demonstrated that above a critical concentration, treatment with all-trans-farnesol induces cell-death in Nicotiana tabacum L. cv Bright Yellow-2 (TBY-2) cells. Now we used a fluorescent analog of farnesol (Fol(FLUO)), in which an isoprene unit is replaced by the fluorochrome 7-nitrobenz-2-oxa-1,3-diazol-4-yl, to visualize how cell integrity is affected. Fol(FLUO) exhibited the same toxicity as the natural compound and was shown to be readily taken up by TBY-2 cells, followed by integration into subcellular membrane structures. Although the plasma membrane seemed not to be labeled, Fol(FLUO) was associated with the tonoplast, endoplasmic reticulum, and Golgi apparatus or lipid bodies. Longer exposure times and increased Fol(FLUO) accumulation triggered the formation and proliferation of new membrane structures of as yet unknown function. Finally, at even higher and clearly cytotoxic concentrations of the analog, the cell contents became clearly disorganized, with cell swelling and ultimately plasmolysis.


Assuntos
Farneseno Álcool/administração & dosagem , Farneseno Álcool/farmacocinética , Nicotiana/efeitos dos fármacos , Nicotiana/metabolismo , Espectrometria de Fluorescência/métodos , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Corantes Fluorescentes , Taxa de Depuração Metabólica , Coloração e Rotulagem/métodos , Nicotiana/citologia
14.
J Cereb Blood Flow Metab ; 23(6): 728-38, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12796721

RESUMO

Traumatic brain injury activates N-methyl-d-aspartate receptors (NMDAR) inducing activation of the Ras protein (a key regulator of cell growth, survival, and death) and its effectors. Thus, trauma-induced increase in active Ras-GTP might contribute to traumatic brain injury pathology. Based on this hypothesis, a new concept of neuroprotection is proposed, examined here by investigating the effect of the Ras inhibitor S-trans, trans-farnesylthiosalicylic acid (FTS) in a mouse model of closed head injury (CHI). Mice subjected to CHI were treated systemically 1 h later with FTS (5 mg/kg) or vehicle. After 1 h, Ras-GTP in the contused hemisphere showed a significant (3.8-fold) increase, which was strongly inhibited by FTS (82% inhibition) or by the NMDA-receptor antagonist MK-801 (53%). Both drugs also decreased active (phosphorylated) extracellular signal-regulated kinase. FTS prevented the CHI-induced reduction in NMDAR binding in cortical, striatal, and hippocampal regions, measured by [3H]-MK-801 autoradiography, and decreased lesion size by 50%. It also reduced CHI-induced neurologic deficits, indicated by the highly significant (P < 0.0001) 60% increase in extent of recovery. Thus, FTS provided long-term neuroprotection after CHI, rescuing NMDAR binding in the contused hemisphere and profoundly reducing neurologic deficits. These findings suggest that nontoxic Ras inhibitors such as FTS may qualify as neuroprotective drugs.


Assuntos
Inibidores Enzimáticos/farmacocinética , Farneseno Álcool/análogos & derivados , Farneseno Álcool/farmacocinética , Traumatismos Cranianos Fechados/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Salicilatos/farmacocinética , Proteínas ras/antagonistas & inibidores , Animais , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/patologia , Modelos Animais de Doenças , Guanosina Trifosfato/metabolismo , Traumatismos Cranianos Fechados/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Recuperação de Função Fisiológica/efeitos dos fármacos
15.
Biochem Biophys Res Commun ; 293(1): 1-6, 2002 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-12054554

RESUMO

Farnesol and the related isoprenoids, geranylgeraniol, geranylgeranyl pyrophosphate, and farnesyl pyrophosphate, are produced in the endoplasmic reticulum of hepatocytes in mammals, and each serve important biological functions. Of these compounds, only farnesol was shown to significantly inhibit rabbit liver microsomal cytochrome P450 enzymes. The observed inhibition appeared to be reversible, and was not strictly competitive, but rather mixed in nature. Of the activities examined, ethoxycoumarin de-ethylase and diclofenac-4-hydroxylase activities were most sensitive to farnesol, with K(I) and K(I)' values between 11 and 40 microM. Caffeine-8-hydroxylation and taxol-6-hydroxylation were not inhibited at all by farnesol. Farnesol appeared to be a P450 substrate, as well as an inhibitor, as indicated by the NADPH-dependent decrease in farnesol concentration in microsomal incubations, and the metabolism was inhibited by CO, which pointed to the involvement of P450 isozymes.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Inibidores Enzimáticos/farmacologia , Farneseno Álcool/farmacologia , Microssomos Hepáticos/enzimologia , Animais , Inibidores das Enzimas do Citocromo P-450 , Diterpenos/farmacologia , Farneseno Álcool/farmacocinética , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Cinética , Coelhos , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Especificidade por Substrato
16.
Pharm Res ; 11(3): 393-7, 1994 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8008705

RESUMO

The diffusion and metabolism of prednisolone 21-farnesylate were investigated in viable skin of the hairless mouse in vitro. The prodrug ester was extensively metabolized in viable skin, while it was stable in the donor and receptor solutions. The rate of appearance of the prodrug and its metabolite prednisolone was markedly influenced by the direction of the skin placed between the in vitro diffusion half-cells. The rate of bioconversion of the prodrug was determined as a function of the distance from the surface of the skin. The prodrug was increasingly metabolized with the distance from the surface of the skin, indicating that the responsible enzymes are enriched in the lower layers of the viable skin. A model with linearly increasing enzyme activity in the viable skin accounts for the in vitro profiles of the diffusion/metabolism of the prodrug in the viable skin of hairless mouse.


Assuntos
Farneseno Álcool/análogos & derivados , Prednisolona/análogos & derivados , Pró-Fármacos/farmacocinética , Absorção Cutânea , Animais , Biotransformação , Cromatografia Líquida de Alta Pressão , Difusão , Farneseno Álcool/metabolismo , Farneseno Álcool/farmacocinética , Feminino , Técnicas In Vitro , Camundongos , Camundongos Pelados , Modelos Biológicos , Prednisolona/metabolismo , Prednisolona/farmacocinética , Pró-Fármacos/metabolismo , Pele/enzimologia , Espectrofotometria Ultravioleta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA