Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Clin Cancer Res ; 25(20): 6035-6043, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31337643

RESUMO

PURPOSE: Iododeoxyuridine (IUdR) is a potent radiosensitizer; however, its clinical utility is limited by dose-limiting systemic toxicities and the need for prolonged continuous infusion. 5-Iodo-2-pyrimidinone-2'-deoxyribose (IPdR) is an oral prodrug of IUdR that, compared with IUdR, is easier to administer and less toxic, with a more favorable therapeutic index in preclinical studies. Here, we report the clinical and pharmacologic results of a first-in-human phase I dose escalation study of IPdR + concurrent radiation therapy (RT) in patients with advanced metastatic gastrointestinal (GI) cancers. PATIENTS AND METHODS: Adult patients with metastatic GI cancers referred for palliative RT to the chest, abdomen, or pelvis were eligible for study. Patients received IPdR orally once every day × 28 days beginning 7 days before the initiation of RT (37.5 Gy in 2.5 Gy × 15 fractions). A 2-part dose escalation scheme was used, pharmacokinetic studies were performed at multiple time points, and all patients were assessed for toxicity and response to Day 56. RESULTS: Nineteen patients were entered on study. Dose-limiting toxicity was encountered at 1,800 mg every day, and the recommended phase II dose is 1,200 mg every day. Pharmacokinetic analyses demonstrated achievable and sustainable levels of plasma IUdR ≥1 µmol/L (levels previously shown to mediate radiosensitization). Two complete, 3 partial, and 9 stable responses were achieved in target lesions. CONCLUSIONS: Administration of IPdR orally every day × 28 days with RT is feasible and tolerable at doses that produce plasma IUdR levels ≥1 µmol/L. These results support the investigation of IPdR + RT in phase II studies.


Assuntos
Quimiorradioterapia/métodos , Neoplasias Gastrointestinais/terapia , Idoxuridina/farmacocinética , Nucleosídeos de Pirimidina/administração & dosagem , Radiossensibilizantes/administração & dosagem , Administração Oral , Adulto , Idoso , Idoso de 80 Anos ou mais , Fracionamento da Dose de Radiação , Estudos de Viabilidade , Feminino , Neoplasias Gastrointestinais/patologia , Humanos , Idoxuridina/administração & dosagem , Idoxuridina/toxicidade , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Pró-Fármacos/administração & dosagem , Pró-Fármacos/farmacocinética , Pró-Fármacos/toxicidade , Nucleosídeos de Pirimidina/farmacocinética , Nucleosídeos de Pirimidina/toxicidade , Radiossensibilizantes/farmacocinética , Radiossensibilizantes/toxicidade , Resultado do Tratamento
2.
J Med Virol ; 89(7): 1255-1264, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-27943311

RESUMO

This prospective, parallel-group, randomized, double-blind, multicenter study compared the efficacy and safety of FV-100 with valacyclovir for reducing pain associated with acute herpes zoster (HZ). Patients, ≥50 years of age, diagnosed with HZ within 72 h of lesion appearance who had HZ-associated pain, were randomized 1:1:1 to a 7-day course of either FV-100 200 mg QD (n = 117), FV-100 400 mg QD (n = 116), or valacyclovir 1000 mg TID (n =117). Efficacy was evaluated on the basis of the burden of illness (BOI; Zoster Brief Pain Inventory scores); incidence and duration of clinically significant pain (CSP); pain scores; incidence and severity of post-herpetic neuralgia (PHN); and times to full lesion crusting and to lesion healing. Safety was evaluated on the basis of adverse event (AE)/SAE profiles, changes in laboratory and vital signs values, and results of electrocardiograms. The burden of illness scores for pain through 30 days were 114.5, 110.3, and 118.0 for FV-100 200 mg, FV-100 400 mg, and valacyclovir 3000 mg, respectively. The incidences of PHN at 90 days for FV-100 200 mg, FV-100 400 mg, and valacyclovir 3000 mg were 17.8%, 12.4%, and 20.2%, respectively. Adverse event and SAE profiles of the two FV-100 and the valacyclovir groups were similar and no untoward signals or trends were evident. These results demonstrate a potential for FV-100 as an antiviral for the treatment of shingles that could both reduce the pain burden of the acute episode and reduce the incidence of PHN compared with available treatments.


Assuntos
Aciclovir/análogos & derivados , Antivirais/uso terapêutico , Herpes Zoster/complicações , Neuralgia Pós-Herpética/prevenção & controle , Dor/tratamento farmacológico , Nucleosídeos de Pirimidina/uso terapêutico , Valina/análogos & derivados , Aciclovir/administração & dosagem , Aciclovir/efeitos adversos , Aciclovir/uso terapêutico , Idoso , Idoso de 80 Anos ou mais , Antivirais/administração & dosagem , Antivirais/efeitos adversos , Efeitos Psicossociais da Doença , Método Duplo-Cego , Feminino , Herpes Zoster/tratamento farmacológico , Herpes Zoster/epidemiologia , Herpes Zoster/virologia , Humanos , Masculino , Pessoa de Meia-Idade , Neuralgia Pós-Herpética/virologia , Manejo da Dor , Estudos Prospectivos , Nucleosídeos de Pirimidina/administração & dosagem , Nucleosídeos de Pirimidina/efeitos adversos , Valaciclovir , Valina/administração & dosagem , Valina/efeitos adversos , Valina/uso terapêutico
3.
Int J Biochem Cell Biol ; 83: 47-55, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27923747

RESUMO

Tumor necrosis factor-related apoptosis-induced ligand (TRAIL) induces apoptosis selectively in cancer cells without affecting the majority of normal human cells. However, hepatocellular carcinoma (HCC) cells often display resistance to TRAIL-induced apoptosis. Ibulocydine (IB) is an isobutyrate ester pro-drug of a novel synthetic Cdk inhibitor that targets Cdk7 and Cdk9. In this study, we show that treatment with subtoxic doses of IB in combination with TRAIL displays potent cytotoxicity in TRAIL-resistant human HCC cells. Combination of IB and TRAIL was found to synergistically induce apoptosis through activation of caspases, which was blocked by a pan-caspase inhibitor (zVAD). Although the expression of Mcl-1 and survivin were reduced by IB plus TRAIL, overexpression of Mcl-1 and survivin did not block the cell death induced by co-treatment. Moreover, overexpression of Bcl-xL did not significantly interfere with the cell death induced by co-treatment of IB and TRAIL. Interestingly, the combination treatment induced cleavage of Bax, which was translocated to mitochondria upon induction of apoptosis. Furthermore, down-regulation of Bax by small interfering RNA effectively reduced the cell death and loss of mitochondrial membrane potential (MMP) caused by co-treatment with IB and TRAIL. Finally, pre-treatment of HCC cells with a calpain inhibitor effectively blocked IB plus TRAIL-induced cleavage of Bax and apoptosis. Collectively, our results demonstrate that IB increases the sensitivity of human HCC cells to TRAIL via mitochondria signaling pathway mediated by calpain-induced cleavage of Bax, suggesting that combined treatment with IB and TRAIL may offer an effective therapeutic strategy for human HCC.


Assuntos
Apoptose/efeitos dos fármacos , Calpaína/metabolismo , Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Pró-Fármacos/farmacologia , Nucleosídeos de Pirimidina/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Proteína X Associada a bcl-2/metabolismo , Apoptose/fisiologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Caspases/metabolismo , Linhagem Celular Tumoral , Sinergismo Farmacológico , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Pró-Fármacos/administração & dosagem , Nucleosídeos de Pirimidina/administração & dosagem , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/administração & dosagem , Regulação para Cima , Proteína X Associada a bcl-2/antagonistas & inibidores , Proteína X Associada a bcl-2/genética , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
4.
Apoptosis ; 21(10): 1144-57, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27421828

RESUMO

Among anti-cancer candidate drugs, TRAIL might be the most specific agent against cancer cells due to its low toxicity to normal cells. Unfortunately, cancer cells usually develop drug resistance to TRAIL, which is a major obstacle for its clinical application. One promising strategy is co-administrating with sensitizer to overcome cancer cells resistance to TRAIL. Clitocine, a natural amino nucleoside purified from wild mushroom, is recently demonstrated that can induce apoptosis in multidrug-resistant human cancer cells by targeting Mcl-1. In the present study,we found that pretreatment with clitocine dramatically enhances TRAIL lethality in its resistant human colon cancer cells by inducing apoptosis. More importantly, combination of clitocine and TRAIL also effectively inhibits xenograft growth and induces tumor cells apoptosis in athymic mice. The disruption of the binding between Mcl-1 and Bak as well as mitochondrial translocation of Bax mediated by clitocine are identified as the key underlying mechanisms, which leading to mitochondrial membrane permeabilization. Enforced exogenous Mcl-1 can effectively attenuate clitocine/TRAIL-induced apoptosis by suppressing the activation of intrinsic apoptotic pathway. Furthermore, clitocine regulates Mcl-1 expression at the posttranslational level as no obvious change is observed on mRNA level and proteasome inhibitor MG132 almost blocks the Mcl-1 suppression by clitocine. In fact, more ubiquitinated Mcl-1 was detected under clitocine treatment. Our findings indicate that clitocine is potentially an effective adjuvant agent in TRAIL-based cancer therapy.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Nucleosídeos de Pirimidina/administração & dosagem , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Antineoplásicos , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/fisiopatologia , Humanos , Camundongos Nus , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Proteólise/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
5.
Cancer Lett ; 355(2): 253-63, 2014 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-25304383

RESUMO

ABT-737 is a novel anti-apoptotic Bcl-2 family protein inhibitor with high affinity to Bcl-2, Bcl-xl and Bcl-w but relatively low affinity to Mcl-1/A1. Therefore, high level Mcl-1 usually confers human tumor cell resistance to ABT-737. At the present study, we observed that clitocine can induce apoptosis in six tested human colon cancer cell lines accompanied by suppression of Mcl-1. More interestingly, clitocine significantly enhances the ABT-737-mediated lethality by inducing apoptosis. At the molecular level we determined Mcl-1 is the potential target through which clitocine can sensitize human colon cancer cells to ABT-737 induced apoptosis. Knocking-down of Mcl-1 is sufficient to increase cancer cell susceptibility to ABT-737 while its over-expression can significantly reverse this susceptibility. We also determined that clitocine may activate Bak by disrupting the interaction between Mcl-1 and Bak to induce mitochondrial membrane permeabilization. Furthermore, silence of Bak with the specific siRNA effectively attenuates the apoptosis induction by co-treatment of clitocine and ABT-737. Finally, clitocine in combination with ABT-737 significantly suppress the xenograft growth in animal model. Collectively, our studies suggest clitocine can induce apoptosis and potentiate ABT-737 lethality in human colon cancer cells by disrupting the interaction of Mcl-1 and Bak to trigger apoptosis.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Compostos de Bifenilo/farmacologia , Neoplasias do Colo/tratamento farmacológico , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Nitrofenóis/farmacologia , Nucleosídeos de Pirimidina/farmacologia , Sulfonamidas/farmacologia , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Animais , Compostos de Bifenilo/administração & dosagem , Células CACO-2 , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/efeitos dos fármacos , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Células HCT116 , Humanos , Camundongos , Camundongos Nus , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Nitrofenóis/administração & dosagem , Piperazinas/administração & dosagem , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Nucleosídeos de Pirimidina/administração & dosagem , Distribuição Aleatória , Sulfonamidas/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Antimicrob Agents Chemother ; 55(8): 3854-60, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21628542

RESUMO

Hepatitis C virus (HCV) infects an estimated 170 million individuals worldwide, and the current standard of care, a combination of pegylated interferon alpha and ribavirin, is efficacious in achieving sustained viral response in ~50% of treated patients. Novel therapies under investigation include the use of nucleoside analog inhibitors of the viral RNA-dependent RNA polymerase. NM283, a 3'-valyl ester prodrug of 2'-C-methylcytidine, has demonstrated antiviral efficacy in HCV-infected patients (N. Afdhal et al., J. Hepatol. 46[Suppl. 1]:S5, 2007; N. Afdhal et al., J. Hepatol. 44[Suppl. 2]:S19, 2006). One approach to increase the antiviral efficacy of 2'-C-methylcytidine is to increase the concentration of the active inhibitory species, the 5'-triphosphate, in infected hepatocytes. HepDirect prodrug technology can increase intracellular concentrations of a nucleoside triphosphate in hepatocytes by introducing the nucleoside monophosphate into the cell, bypassing the initial kinase step that is often rate limiting. Screening for 2'-C-methylcytidine triphosphate levels in rat liver after oral dosing identified 1-[3,5-difluorophenyl]-1,3-propandiol as an efficient prodrug modification. To determine antiviral efficacy in vivo, the prodrug was administered separately via oral and intravenous dosing to two HCV-infected chimpanzees. Circulating viral loads declined by ~1.4 log(10) IU/ml and by >3.6 log(10) IU/ml after oral and intravenous dosing, respectively. The viral loads rebounded after the end of dosing to predose levels. The results indicate that a robust antiviral response can be achieved upon administration of the prodrug.


Assuntos
Citidina/análogos & derivados , Hepacivirus/efeitos dos fármacos , Hepatite C/tratamento farmacológico , Pró-Fármacos/administração & dosagem , Animais , Antivirais/administração & dosagem , Antivirais/farmacologia , Antivirais/uso terapêutico , Citidina/administração & dosagem , Citidina/farmacologia , Citidina/uso terapêutico , Monofosfato de Citidina/administração & dosagem , Monofosfato de Citidina/análogos & derivados , Monofosfato de Citidina/farmacologia , Monofosfato de Citidina/uso terapêutico , Feminino , Hepatite C/virologia , Hepatócitos/metabolismo , Macaca mulatta , Masculino , Pan troglodytes , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Nucleosídeos de Pirimidina/administração & dosagem , Nucleosídeos de Pirimidina/farmacologia , Nucleosídeos de Pirimidina/uso terapêutico , Ratos , Ratos Sprague-Dawley , Carga Viral/efeitos dos fármacos
7.
Antimicrob Agents Chemother ; 55(6): 2847-54, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21444712

RESUMO

FV-100 is the prodrug of the highly potent anti-varicella zoster virus bicyclic nucleoside analogue CF-1743. To characterize the pharmacokinetics and safety of oral FV-100, 3 randomized, double-blind, placebo-controlled clinical trials were conducted: (i) a single-ascending-dose study in 32 healthy subjects aged 18 to 55 years (100-, 200-, 400-, and 800-mg doses) with an evaluation of the food effect in the 400-mg group; (ii) a multiple-ascending-dose study in 48 subjects aged 18 to 55 years (100 mg once daily [QD], 200 mg QD, 400 mg QD, 400 mg twice a day, and 800 mg QD for 7 days); and (iii) a 2-part study in subjects aged 65 years and older with a single 400-mg dose in 15 subjects and a 400-mg QD dosing regimen for 7 days in 12 subjects. FV-100 was rapidly and extensively converted to CF-1743, the concentration of which remained above that required to reduce viral activity by 50% for the 24-hour dosing period. Renal excretion of CF-1743 was very low. A high-fat meal reduced exposure to CF-1743; a low-fat meal did not. Pharmacokinetic parameters for the elderly subjects were comparable to those for the younger subjects. FV-100 was well tolerated by all subjects. The pharmacokinetic and safety profiles of FV-100 support its continued investigation for the treatment of herpes zoster and prevention of postherpetic neuralgia with once-daily dosing and without dose modifications for elderly or renally impaired patients.


Assuntos
Antivirais/farmacocinética , Herpes Zoster/tratamento farmacológico , Pró-Fármacos/farmacocinética , Nucleosídeos de Pirimidina/farmacocinética , Administração Oral , Adolescente , Adulto , Fatores Etários , Idoso , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Nucleosídeos de Pirimidina/administração & dosagem , Nucleosídeos de Pirimidina/efeitos adversos , Nucleosídeos de Pirimidina/sangue
8.
Bioorg Med Chem Lett ; 19(22): 6264-7, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19833513

RESUMO

We report the preparation of 2'-alpha-F, 2'-beta-F and 2',2'-difluoro analogues of the leading anti-varicella zoster virus (VZV) pentylphenyl BCNA Cf 1743. VZV thymidine kinase showed the highest phosphorylating capacity for the beta-fluoro derivative, that retained equal antiviral potency as the parent compound. In contrast, the alpha-fluoro- and 2',2'-difluoro BCNA derivatives were markedly less (approximately 100-fold) antivirally active.


Assuntos
Antivirais/farmacologia , Desenho de Fármacos , Herpesvirus Humano 3/efeitos dos fármacos , Nucleosídeos/farmacologia , Nucleosídeos de Pirimidina/farmacologia , Replicação Viral/efeitos dos fármacos , Compostos Bicíclicos com Pontes/química , Compostos Bicíclicos com Pontes/farmacologia , Bromodesoxiuridina/química , Bromodesoxiuridina/farmacologia , Linhagem Celular , Interações Medicamentosas , Fluoretos/química , Células HeLa , Herpesvirus Humano 3/metabolismo , Humanos , Testes de Sensibilidade Microbiana , Modelos Moleculares , Nucleosídeos/química , Nucleosídeos de Pirimidina/administração & dosagem , Relação Estrutura-Atividade , Especificidade por Substrato , Timidina Quinase/metabolismo , Replicação Viral/fisiologia
9.
J Sep Sci ; 32(9): 1275-83, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19347863

RESUMO

A very accurate and selective LC-MS/MS method was developed and validated for the quantification of 2'-C-modified nucleoside triphosphate in liver tissue samples. An efficient pretreatment procedure of liver tissue samples was developed, using a fully automated SPE procedure with 96-well SPE plate (weak anion exchange sorbent, 30 mg). Nucleotide hydrophilic interaction chromatography has been performed on an aminopropyl column (100 mm x 2.0 mm, 3 microm) using a gradient mixture of ACN and ACN/water (5:95 v/v) with 20 mM ammonium acetate at pH 9.45 as mobile phase at 300 microL/min flow rate. The 2'-C-modified nucleoside triphosphate was detected in the negative ESI mode in multiple reaction monitoring (MRM) mode. Calibration curve was linear over the 0.05-50 microM concentration range. Satisfying results, confirming the high reliability of the established LC-MS/MS method, were obtained for intraday precision (CV = 2.5-9.1%) and accuracy (92.6-94.8%) and interday precision (CV = 9.6-11.5%) and accuracy (94.4-102.4%) as well as for recovery (82.0-112.6%) and selectivity. The method has been successfully applied for pharmacokinetic studies of 2'-C-methyl-cytidine-triphosphate in liver tissue samples.


Assuntos
Citidina Trifosfato/análogos & derivados , Citidina Trifosfato/análise , Fígado/química , Espectrometria de Massas em Tandem/métodos , Animais , Antivirais/administração & dosagem , Antivirais/metabolismo , Calibragem , Cromatografia Líquida/métodos , Masculino , Pró-Fármacos/administração & dosagem , Pró-Fármacos/metabolismo , Nucleosídeos de Pirimidina/administração & dosagem , Nucleosídeos de Pirimidina/metabolismo , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Extração em Fase Sólida/métodos , Solventes/química
10.
Best Pract Res Clin Gastroenterol ; 22(6): 1123-36, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19187871

RESUMO

Specifically Targeted Antiviral Therapy against hepatitis C virus (STAT-C) stands for a new era in the treatment of patients with chronic hepatitis C. Results from recent trials with protease and polymerase inhibitors indicate that therapy with a single HCV specific compound will not be sufficient to eradicate hepatitis C virus infection and that combination therapy will be necessary to improve sustained virologic response rates. The search for the optimal combination of STAT-C compounds with peginterferon alfa with or without ribavirin is currently under investigation in several clinical trials. Overall the current studies indicate that peginterferon alfa and ribavirin remain the backbone of antiviral therapy of chronic hepatitis C even in the era of STAT-C. Nevertheless, it can be anticipated that combination of STAT-C compounds with non-overlapping resistance profiles could improve response to antiviral therapy. Promising combinations are protease inhibitors plus nucleoside analogue and non-nucleoside analogue polymerase inhibitors.


Assuntos
Antivirais/administração & dosagem , Sistemas de Liberação de Medicamentos/tendências , Hepatite C Crônica/tratamento farmacológico , Citidina/administração & dosagem , Citidina/análogos & derivados , RNA Polimerases Dirigidas por DNA/administração & dosagem , Farmacorresistência Viral , Quimioterapia Combinada , Previsões , Humanos , Interferon alfa-2 , Interferon-alfa/administração & dosagem , Nucleosídeos/administração & dosagem , Oligopeptídeos/administração & dosagem , Polietilenoglicóis/administração & dosagem , Pró-Fármacos/administração & dosagem , Prolina/administração & dosagem , Prolina/análogos & derivados , Inibidores de Proteases/administração & dosagem , Nucleosídeos de Pirimidina/administração & dosagem , Proteínas Recombinantes , Ribavirina/administração & dosagem
11.
Expert Opin Investig Drugs ; 16(9): 1415-24, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17714027

RESUMO

IPdR (5-iodo-2-pyrimidinone-2'-deoxyribose) is a novel orally available, halogenated thymidine (TdR) analog and is a potential radiosensitizer for use in human tumors, such as rectal, pancreas, sarcoma and glioma tumors. IPdR is a prodrug that is efficiently converted to IUdR (5-iodo-2'-deoxyuridine), an intravenous radiosensitizer by a hepatic aldehyde oxidase, resulting in high IPdR and IUdR plasma levels in mice for > or = 1 h after oral IPdR. Athymic mice tolerated oral IPdR to doses up to 1500 mg/kg/day t.i.d. for 6 - 14 days without significant systemic toxicities. A number of in vivo preclinical studies have demonstrated that IPdR is a superior radiosensitizer compared with IUdR given as a continuous infusion in terms of safety and efficacy with a significantly lower toxicity profile, including gastrointestinal and hematologic side effects. A preclinical study has shown that IPdR is effective in inducing human colon cancer xenograft radiosensitization in drug-resistant DNA mismatch repair-proficient and -deficient tumor models, as well as in human globlastoma xenograft. In anticipation of performing a clinical Phase I trial in humans, investigators also studied the drug pharmacokinetics and host toxicities in two non-rodent, animal species during a 14-day treatment course. Dose-limiting systemic toxicities (diarrhea, emesis, weight loss and decreased motor activity) were observed in ferrets receiving IPdR at 1500 mg/kg/day on a 14-day schedule that were not found previously in athymic mice. Recently, a once-daily IPdR dosing up to 2000/mg/kg for 28 days in Fischer-344 rats showed reversible mild-to-moderate systemic toxicities without any severe or life-threatening toxicities. However, in all preclinical toxicity studies so far, no significant hematologic, biochemical or histopathologic changes have been found. Hepatic aldehyde oxidase activity was reduced in a dose-dependent fashion in the ferret liver, suggesting partial enzyme inactivation by this IPdwR schedule, but that is not found in Fischer-344 rats. The plasma pharmacokinetic profile in Rhesus monkeys showing biexponential clearance are similar to previously published data in athymic mice. In this paper, the authors review the development, mechanism of action, preclinical data and rationale for clinical studies.


Assuntos
Drogas em Investigação/administração & dosagem , Nucleosídeos de Pirimidina/administração & dosagem , Radiossensibilizantes/administração & dosagem , Animais , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos/métodos , Drogas em Investigação/farmacocinética , Humanos , Nucleosídeos de Pirimidina/farmacocinética , Radiossensibilizantes/farmacocinética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
12.
Biopharm Drug Dispos ; 28(4): 187-97, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17377948

RESUMO

The pharmacokinetics of L-FMAUS after intravenous and oral administration (20, 50 and 100 mg/kg) to rats, gastrointestinal first-pass effect of L-FMAUS (50 mg/kg) in rats, in vitro stability of L-FMAUS, blood partition of L-FMAUS between plasma and blood cells of rat blood, and protein binding of L-FMAUS to 4% human serum albumin were evaluated. L-FMAUS is being evaluated in a preclinical study as a novel antiviral agent. Although the dose-normalized AUC values of L-FMAUS were not significantly different among the three doses after intravenous and oral administration, no trend was apparent between the dose and dose-normalized AUC. After oral administration of L-FMAUS (50 mg/kg), approximately 2.37% of the oral dose was not absorbed, and the extent of absolute oral bioavailability (F) was approximately 11.5%. The gastrointestinal first-pass effect was approximately 85% of the oral dose. The first-pass effects of L-FMAUS in the lung, heart and liver were almost negligible, if any, in rats. Hence, the small F of L-FMAUS in rats was mainly due to the considerable gastrointestinal first-pass effect. L-FMAUS was stable in rat gastric juices. The plasma-to-blood cells partition ratio of L-FMAUS was 2.17 in rat blood. The plasma protein binding of L-FMAUS in rats was 98.6%.


Assuntos
Antivirais/farmacocinética , Nucleosídeos de Pirimidina/farmacocinética , Administração Oral , Animais , Antivirais/administração & dosagem , Antivirais/farmacologia , Área Sob a Curva , Disponibilidade Biológica , Estabilidade de Medicamentos , Meia-Vida , Infusões Intravenosas , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Masculino , Nucleosídeos de Pirimidina/administração & dosagem , Nucleosídeos de Pirimidina/farmacologia , Ratos , Ratos Sprague-Dawley
13.
Clin Cancer Res ; 11(20): 7499-507, 2005 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16243824

RESUMO

PURPOSE: 5-Iodo-2-pyrimidinone-2'-deoxyribose (IPdR) is an oral prodrug of 5-iodo-2'-deoxyuridine (IUdR), an in vitro/in vivo radiosensitizer. IPdR can be rapidly converted to IUdR by a hepatic aldehyde oxidase. Previously, we found that the enzymatic conversion of IPdR to IUdR could be transiently reduced using a once daily (q.d.) treatment schedule and this may affect IPdR-mediated tumor radiosensitization. The purpose of this study is to measure the effect of different drug dosing schedules on tumor radiosensitization and therapeutic index in human glioblastoma xenografts. EXPERIMENTAL DESIGN: Three different IPdR treatment schedules (thrice a day, t.i.d.; every other day, q.o.d.; every 3rd day, q.3.d.), compared with a q.d. schedule, were analyzed using athymic nude mice with human glioblastoma (U251) s.c. xenografts. Plasma pharmacokinetics, IUdR-DNA incorporation in tumor and normal proliferating tissues, tumor growth delay following irradiation, and body weight loss were used as end points. RESULTS: The t.i.d. schedule with the same total daily doses as the q.d. schedule (250, 500, or 1,000 mg/kg/d) improved the efficiency of IPdR conversion to IUdR. As a result, the percentage of IUdR-DNA incorporation was higher using the t.i.d. schedule in the tumor xenografts as well as in normal small intestine and bone marrow. Using a fixed dose (500 mg/kg) per administration, the q.o.d. and q.3.d. schedules also showed greater IPdR conversion than the q.d. schedule, related to a greater recovery of hepatic aldehyde oxidase activity prior to the next drug dosing. In the tumor regrowth assay, all IPdR treatment schedules showed significant increases of regrowth delays compared with the control without IPdR (q.o.d., 29.4 days; q.d., 29.7 days; t.i.d., 34.7 days; radiotherapy alone, 15.7 days). The t.i.d. schedule also showed a significantly enhanced tumor growth delay compared with the q.d. schedule. Additionally, the q.o.d. schedule resulted in a significant reduction in systemic toxicity. CONCLUSIONS: The t.i.d. and q.o.d. dosing schedules improved the efficiency of enzymatic activation of IPdR to IUdR during treatment and changed the extent of tumor radiosensitization and/or systemic toxicity compared with a q.d. dosing schedule. These dosing schedules will be considered for future clinical trials of IPdR-mediated human tumor radiosensitization.


Assuntos
Glioblastoma/tratamento farmacológico , Nucleosídeos de Pirimidina/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Administração Oral , Animais , Área Sob a Curva , DNA de Neoplasias/metabolismo , Relação Dose-Resposta a Droga , Esquema de Medicação , Glioblastoma/patologia , Glioblastoma/radioterapia , Humanos , Idoxuridina/sangue , Idoxuridina/metabolismo , Camundongos , Camundongos Nus , Nucleosídeos de Pirimidina/farmacocinética , Nucleosídeos de Pirimidina/uso terapêutico , Radiossensibilizantes/administração & dosagem , Radiossensibilizantes/farmacocinética , Radiossensibilizantes/uso terapêutico , Fatores de Tempo
14.
Pharm Res ; 21(6): 914-9, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15212153

RESUMO

PURPOSE: To determine the in-vitro dermal delivery of a new class of lipophilic, highly potent and uniquely selective anti-VZV nucleoside analogues in comparison with aciclovir. METHODS: Three test compounds (Cf1698, Cf1743, Cf1712) and aciclovir were formulated into propylene glycol/aqueous cream BP formulations and finite doses applied to full-thickness pig ear skin for 48 hours in vertical Franz-type diffusion cells. Receptor phase samples were taken at specific intervals to determine permeation, and depth profiles were constructed following tape stripping and membrane separation. RESULTS: All three test compounds reached the target basal epidermis in concentrations suggesting they would be highly efficacious in reducing viral load. Furthermore, the data showed that each of the test compounds would perform in a far superior manner to aciclovir, the current treatment of choice. CONCLUSIONS: The dermatomal site of viral replication during secondary infection--the basal epidermis--was successfully targeted. Topical delivery of these compounds is highly promising as a new first line treatment of VZV infections. By attacking the virus at the first sign of reactivation, it is proposed that the extent of damage caused by the virus would be significantly lowered, thereby limiting the extent and severity of post-herpetic neuralgia.


Assuntos
Antivirais/farmacocinética , Sistemas de Liberação de Medicamentos/métodos , Herpesvirus Humano 3/efeitos dos fármacos , Nucleosídeos/farmacocinética , Pele/efeitos dos fármacos , Aciclovir/administração & dosagem , Aciclovir/farmacocinética , Administração Cutânea , Animais , Antivirais/administração & dosagem , Antivirais/síntese química , Química Farmacêutica/métodos , Herpesvirus Humano 3/crescimento & desenvolvimento , Nucleosídeos/administração & dosagem , Nucleosídeos/síntese química , Pomadas/administração & dosagem , Pomadas/química , Pomadas/farmacocinética , Permeabilidade , Soluções Farmacêuticas/administração & dosagem , Soluções Farmacêuticas/farmacocinética , Nucleosídeos de Pirimidina/administração & dosagem , Nucleosídeos de Pirimidina/síntese química , Nucleosídeos de Pirimidina/farmacocinética , Pele/metabolismo , Absorção Cutânea/efeitos dos fármacos , Suspensões/administração & dosagem , Suspensões/química , Suspensões/farmacocinética , Suínos , Reino Unido
15.
J Natl Cancer Inst ; 95(5): 399-409, 2003 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-12618505

RESUMO

BACKGROUND: Gene silencing by abnormal methylation of promoter regions of regulatory genes is commonly associated with cancer. Silenced tumor suppressor genes are obvious targets for reactivation by methylation inhibitors such as 5-azacytidine (5-Aza-CR) and 5-aza-2'-deoxycytidine (5-Aza-CdR). However, both compounds are chemically unstable and toxic and neither can be given orally. We characterized a new demethylating agent, zebularine [1-(beta-D-ribofuranosyl)-1,2-dihydropyrimidin-2-one], which is a chemically stable cytidine analog. METHODS: We tested the ability of zebularine to reactivate a silenced Neurospora crassa gene using a hygromycin gene reactivation assay. We then analyzed the ability of zebularine to inhibit DNA methylation in C3H 10T1/2 Cl8 (10T1/2) mouse embryo cells as assayed by induction of a myogenic phenotype and in T24 human bladder carcinoma cells, using the methylation-sensitive single nucleotide primer extension (Ms-SNuPE) assay. We also evaluated the effects of zebularine (administered orally or intraperitoneally) on growth of EJ6 human bladder carcinoma cells grown in BALB/c nu/nu mice (five mice per group) and the in vivo reactivation of a methylated p16 gene in these cells. All statistical tests were two-sided. RESULTS: In N. crassa, zebularine inhibited DNA methylation and reactivated a gene previously silenced by methylation. Zebularine induced the myogenic phenotype in 10T1/2 cells, which is a phenomenon unique to DNA methylation inhibitors. Zebularine reactivated a silenced p16 gene and demethylated its promoter region in T24 bladder carcinoma cells in vitro and in tumors grown in mice. Zebularine was only slightly cytotoxic to T24 cells in vitro (1 mM zebularine for 48 hours decreased plating efficiency by 17% [95% confidence interval (CI) = 12.8% to 21.2%]) and to tumor-bearing mice (average maximal weight change in mice treated with 1000 mg/kg zebularine = 11% [95% CI = 4% to 19%]). Compared with those in control mice, tumor volumes were statistically significantly reduced in mice treated with high-dose zebularine administered by intraperitoneal injection (P<.001) or by oral gavage (P<.001). CONCLUSIONS: Zebularine is a stable DNA demethylating agent and the first drug in its class able to reactivate an epigenetically silenced gene by oral administration.


Assuntos
Cinamatos , Metilação de DNA/efeitos dos fármacos , DNA de Neoplasias/metabolismo , Inativação Gênica , Higromicina B/análogos & derivados , Neurospora crassa/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Nucleosídeos de Pirimidina/farmacologia , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/metabolismo , Administração Oral , Animais , Southern Blotting , Citidina/análogos & derivados , DNA de Neoplasias/efeitos dos fármacos , Relação Dose-Resposta a Droga , Esquema de Medicação , Embrião de Mamíferos , Regulação Fúngica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Humanos , Higromicina B/metabolismo , Infusões Parenterais , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neurospora crassa/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/efeitos dos fármacos , Nucleosídeos de Pirimidina/administração & dosagem , Nucleosídeos de Pirimidina/química , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Células Tumorais Cultivadas , Neoplasias da Bexiga Urinária/genética
16.
Cancer Res ; 54(10): 2695-700, 1994 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-8168099

RESUMO

5-iodo-2-pyrimidinone-2'-deoxyribose (IPdR) was recently reported to be converted to 5-iodo-2'-deoxyuridine (IUdR) by an aldehyde oxidase, most concentrated in liver tissue. We questioned whether IPdR could be used as a p.o. hepatotropic prodrug to increase the percentage of IUdR-DNA incorporation into liver tumors compared to normal liver with acceptable systemic toxicity. Athymic nude mice with human colon cancer (HCT-116) xenograft tumors as liver metastases and s.c. flank tumors received daily p.o. boluses (via gastric tubes) of IUdR or IPdR for 6 days. The maximum tolerated dose of IUdR was 250 mg/kg/day and was associated with a > 10% weight loss and a high percentage of IUdR-DNA incorporation (> 5%) into normal bone marrow and intestine. In contrast, animals tolerated escalating doses of IPdR to 1 gm/kg/day without weight loss and with less (1.5-4%) IUdR-DNA incorporation in normal tissues. Pharmacokinetic analysis of p.o. IPdR showed peak plasma levels of IPdR and IUdR within 15-45 min, suggesting efficient conversion of IPdR to IUdR. Aldehyde oxidase activity was found in normal liver tissue but not in other normal or tumor tissues. Additionally, we found a 2-3 times greater percentage of IUdR-DNA incorporation in tumor with IPdR than IUdR at the highest doses used. However, no differential effect in the percentage of IUdR-DNA incorporation was noted between liver metastases and s.c. tumors with either IPdR or IUdR. We conclude that p.o. IPdR offers a greater therapeutic index for tumor incorporation (and presumably radiosensitization) than a similar schedule of IUdR.


Assuntos
DNA/metabolismo , Idoxuridina/farmacocinética , Neoplasias Hepáticas/metabolismo , Fígado/metabolismo , Nucleosídeos de Pirimidina/farmacocinética , Administração Oral , Animais , Neoplasias do Colo/metabolismo , Feminino , Humanos , Idoxuridina/administração & dosagem , Idoxuridina/toxicidade , Neoplasias Hepáticas/secundário , Camundongos , Transplante de Neoplasias , Nucleosídeos de Pirimidina/administração & dosagem , Nucleosídeos de Pirimidina/toxicidade , Distribuição Tecidual
17.
Nucleic Acids Symp Ser ; (29): 51-2, 1993.
Artigo em Inglês | MEDLINE | ID: mdl-8247791

RESUMO

N3-Substituted pyrimidine nucleoside and their related compounds were synthesized. The central nervous system (CNS) depressant effect of the compounds such as hypnotic activity and synergistic effect with pentobarbital have been evaluated. N3-Phenacyl derivatives of uridine (2), thymidine (3), deoxyuridine (4), 6-azauridine (5) and arabinofuranosyluracil (6) exhibited the hypnotic activity, whereas N3-phenacyl-2',3'-O-isopropylideneuridine did not. Compound 2, 3, 4 and 6 significantly enhanced pentobarbital-induced sleep. The results indicate that CNS depressant effect of pyrimidine nucleoside derivatives might relate to functional group at the N3-position on oxopyrimidine ring and stereospecificity of sugar moiety at the N1-position.


Assuntos
Hipnóticos e Sedativos/farmacologia , Nucleosídeos de Pirimidina/farmacologia , Animais , Sistema Nervoso Central/efeitos dos fármacos , Hipnóticos e Sedativos/administração & dosagem , Hipnóticos e Sedativos/química , Camundongos , Estrutura Molecular , Nucleosídeos de Pirimidina/administração & dosagem , Nucleosídeos de Pirimidina/química
18.
Ann R Coll Surg Engl ; 69(1): 36-8, 1987 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-3566116

RESUMO

Cytotoxic liver perfusion via the portal vein has been used with some success as an adjuvant to surgery for colorectal carcinoma. We have used an alternative method of cannulating the portal vein at the time of operation and support the use of this method as well as the use of short-term 5FU infusion via the portal vein as being safe and of low systemic toxicity.


Assuntos
Antineoplásicos/administração & dosagem , Cateterismo/métodos , Neoplasias Hepáticas/secundário , Veia Porta , Adenocarcinoma/cirurgia , Neoplasias do Colo/cirurgia , Fluoruracila/administração & dosagem , Fluoruracila/análogos & derivados , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Nucleosídeos de Pirimidina/administração & dosagem , Neoplasias Retais/cirurgia
19.
Gan To Kagaku Ryoho ; 11(9): 1848-54, 1984 Sep.
Artigo em Japonês | MEDLINE | ID: mdl-6433807

RESUMO

FU-O-G, the O-glucuronide methylester of 5-fluorouracil (5-FU), is a compound with a unique chemical structure. It is an antitumor agent of the prodrug type which exerts its activity by enzymatically liberating 5-FU in tumor tissues. It has been reported that the antitumor activity of this compound is superior to those of 5-FU and Tegafur (FT-207) in the treatment of various transplantable tumors. In this study, long-term administration of FU-O-G to mice bearing relatively slow-growing tumors such as Lewis lung carcinoma, mammary tumor FM3 A and hepatoma MH-134 was carried out. Consequently, FU-O-G was shown to be remarkably effective against those tumors, whereas 5-FU and FT-207 were hardly effective. Long-term daily administration was shown to be more effective than intermittent dosage in the treatment of Lewis lung carcinoma. In combination therapies of FU-O-G with other antitumor drugs, FU-O-G exhibited a synergistic effect against Lewis lung carcinoma when combined with Carboquone (CQ) or Nimustine hydrochloride (ACNU).


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Nucleosídeos de Pirimidina/uso terapêutico , Animais , Carbazilquinona/administração & dosagem , Esquema de Medicação , Sinergismo Farmacológico , Feminino , Fluoruracila/administração & dosagem , Neoplasias Hepáticas Experimentais/mortalidade , Neoplasias Pulmonares/mortalidade , Camundongos , Transplante de Neoplasias , Nimustina , Compostos de Nitrosoureia/administração & dosagem , Nucleosídeos de Pirimidina/administração & dosagem , Tegafur/administração & dosagem
20.
Gan ; 72(2): 220-5, 1981 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-6793441

RESUMO

The antitumor activity of a glucuronide of 5-fluorouracil, methyl (5-fluoro-1H-2-oxo-4-pyrimidinyl beta-D-glucopyranosid) uronate (FU-O-G), which is a 5-fluorouracil (5-FU) derivative with remarkably low toxicity, was studied. The antitumor activity of this compound was superior to those of 5-FU and 1-(2-tetrahydrofuryl)-5-fluorouracil (Tegafur, Ftorafur) in the treatment of transplantable tumors, not only 5-FU-sensitive tumors such as adenocarcinoma 755, lymphosarcoma LS-1, and plasmacytoma X5563, but also 5-FU-resistant tumors such as Lewis lung carcinoma and Walker carcinosarcoma 256. Furthermore, in the treatment of Lewis lung carcinoma, which responds poorly to 5-FU and Ftorafur, daily administration of FU-O-G at a dose of 400 mg/kg for 30 days produced a 92% increase in life span without marked loss of body weight, though short-term administration (such as 5 days) was barely effective. Thus, it appears that FU-O-G is an antitumor agent suitable for long-term administration. These findings correspond with the results of an enzymological study which showed selective activation of FU-O-G by beta-glucosidase in tumor cells and indicated that the compound is a marked form of 5-FU.


Assuntos
Antineoplásicos/farmacologia , Nucleosídeos de Pirimidina/farmacologia , Animais , Carcinoma/tratamento farmacológico , Carcinoma 256 de Walker/tratamento farmacológico , Feminino , Fluoruracila/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos , Neoplasias Experimentais/tratamento farmacológico , Nucleosídeos de Pirimidina/administração & dosagem , Ratos , Tegafur/farmacologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA