Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cancer Chemother Pharmacol ; 63(3): 441-50, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18414865

RESUMO

PURPOSE: To determine the maximum tolerated dose (MTD) and dose-limiting toxicity (DLT) of the combination of weekly oxaliplatin x 4, weekly irinotecan x 4 and capecitabine Monday through Friday for 4 weeks of every 6 week cycle in patients with solid tumors; to determine the pharmacokinetic profile of these agents in this combination; to observe patients for clinical anti-tumor response. METHODS: Twenty-two patients with metastatic solid tumors received oxaliplatin 60 mg/m(2) weekly x 4, irinotecan beginning at a dose of 40 mg/m(2) weekly x 4, and capecitabine Monday through Friday for 4 weeks of every 6 week cycle, initially at 1,000 mg twice daily (bid). RESULTS: The MTD was oxaliplatin 60 mg/m(2) weekly x 4, irinotecan 50 mg/m(2) weekly x 4 and capecitabine 450 mg bid Monday through Friday for 4 weeks of every 6 week cycle. One of six patients at this dose level developed DLT of nausea, vomiting, and diarrhea. Among patients treated with a constant capecitabine dose of 450 mg bid, there was a higher mean AUC of 5-FU in women than in men (mean +/- SD: 892 +/- 287 nM h vs. 537 +/- 182 nM h; Mann-Whitney two-tailed, P = 0.02). There was one complete response in a patient with gastric cancer. CONCLUSION: The novel schedule of weekly oxaliplatin, weekly irinotecan, and capecitabine Monday through Friday, all administered for 4 weeks of every 6 week cycle, evaluated in this phase I trial is well-tolerated and demonstrated activity in a patient with gastric cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias/tratamento farmacológico , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Área Sob a Curva , Camptotecina/administração & dosagem , Camptotecina/análogos & derivados , Capecitabina , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Feminino , Fluoruracila/administração & dosagem , Fluoruracila/análogos & derivados , Humanos , Irinotecano , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Compostos Organoplatínicos/administração & dosagem , Oxaliplatina
2.
Cancer Chemother Pharmacol ; 62(4): 647-53, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18040687

RESUMO

BACKGROUND: The pyrimidine analogue gemcitabine (2', 2'-difluorodeoxycitidine, dFdC) is active against pancreatic cancer, and its high clearance (CL(tb)) and low incidence of local toxicity make it an excellent candidate for evaluation as intraperitoneal (IP) therapy. We designed a dosing schema that used multiple sequential exchanges of a peritoneal dialysate containing dFdC in an effort to produce prolonged IP dFdC exposure. METHODS: As part of a study involving multi-modality therapy for advanced pancreatic adenocarcinoma, patients were treated with four 6-h IP dwells of dFdC (50 mg/m(2) in 2 l) over a 24-h period. A second 24-h cycle of IP dFdC therapy was repeated 1 week later. Each exchange of dialysate contained 50 mg/m(2) dFdC in 2 l of commercial 1.5% dextrose dialysis solution. Plasma and peritoneal fluid were analyzed by HPLC to determine concentrations of dFdC and its inactive metabolite 2', 2' difluorodeoxyuridine (dFdU). Clinical data were recorded to note drug toxicity and response. RESULTS: Nine patients underwent IP dFdC therapy, and eight were able to receive two cycles. There were no recorded significant toxicities. Low plasma dFdC concentrations (<1 microg/ml) were present transiently in seven of nine patients, and dFdC was not detectable in the plasma of the other two. Plasma dFdU concentrations were low but increased gradually until 12 h and then declined little if any. IP dFdC concentrations declined rapidly, and dFdC was seldom measurable prior to administration of the next scheduled 6-h dwell. dFdU concentrations in peritoneal fluid were very low (<0.5 microg/ml) throughout treatment. The mean area under the concentration versus time curve (AUC) for dFdC in peritoneal fluid was 182 microg/ml x h, which was approximately 70x the AUC of dFdC reported in the ascites of a patient undergoing systemic dFdC therapy. CONCLUSIONS: IP dFdC was well tolerated, and no significant toxicities were noted. The rapid decrease in peritoneal dFdC concentrations and low concentrations of IP dFdU imply almost total absorption of IP-administered dFdC. Little, if any, dFdC could be detected in plasma, but the steady-state plasma dFdU concentrations also imply absorption and inactivation of virtually all IP-administered dFdC. These findings are consistent with the known high CL(tb) and low incidence of local toxicity of dFdC and argue for its further evaluation as a drug for IP therapy.


Assuntos
Adenocarcinoma/metabolismo , Antimetabólitos Antineoplásicos/farmacocinética , Desoxicitidina/análogos & derivados , Neoplasias Pancreáticas/metabolismo , Adenocarcinoma/mortalidade , Adenocarcinoma/terapia , Adulto , Antimetabólitos Antineoplásicos/análise , Antimetabólitos Antineoplásicos/uso terapêutico , Líquido Ascítico/química , Cromatografia Líquida de Alta Pressão , Terapia Combinada , Desoxicitidina/análise , Desoxicitidina/farmacocinética , Desoxicitidina/uso terapêutico , Soluções para Diálise/química , Feminino , Humanos , Injeções Intraperitoneais , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/terapia , Diálise Peritoneal , Projetos Piloto , Taxa de Sobrevida , Gencitabina
3.
Clin Cancer Res ; 13(7): 2121-7, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17404095

RESUMO

PURPOSE: Interaction of 17-allylamino-17-demethoxygeldanamycin (17-AAG) with heat shock protein 90 results in proteasomal degradation of many proteins, including Her-2-neu, with subsequent decreased expression of insulin-like growth factor binding protein-2 (IGFBP-2). Concentrations of both IGFBP-2 and Her-2 extracellular domain (Her-2 ECD) in sera of mice bearing BT474 human breast cancer xenografts decrease after 17-AAG treatment. We investigated whether this phenomenon occurred in patients. MATERIALS AND METHODS: Eight to 15 plasma samples were obtained between 0 and 72 h from 27 patients treated with single-agent 17-AAG at doses between 10 and 307 mg/m(2) and 18 patients treated with 17-AAG at doses between 220 and 450 mg/m(2) combined with 70 to 75 mg/m(2) of docetaxel. Pretreatment plasma samples were also obtained from 12 healthy volunteers. Plasma IGFBP-2 and Her-2 ECD concentrations were quantitated by ELISA. RESULTS: Pretreatment plasma IGFBP-2 concentrations in patients (171 +/- 116 ng/mL) were 2-fold higher than those in healthy volunteers (85 +/- 44 ng/mL; P < 0.05). Following 17-AAG treatment, there were no consistent dose-dependent or time-dependent changes in plasma IGFBP-2 and Her-2 ECD concentrations. IGFBP-2 concentrations decreased by >or=40% in 8 patients, increased 2- to 5-fold in 8 patients, and remained essentially unchanged in 29 patients. Her-2 ECD concentrations decreased by >or=40% in 10 patients, increased 1.5- to 5-fold in 2 patients, and remained essentially unchanged in 25 patients. CONCLUSIONS: As previously reported, IGFBP-2 concentrations in plasma of cancer patients are significantly higher than those in healthy volunteers. In contrast to a mouse model, 17-AAG treatment was not consistently associated with decreases in IGFBP-2 or Her-2 ECD concentrations in patient plasma.


Assuntos
Antineoplásicos/uso terapêutico , Benzoquinonas/uso terapêutico , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/sangue , Lactamas Macrocíclicas/uso terapêutico , Neoplasias/tratamento farmacológico , Receptor ErbB-2/sangue , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores Tumorais/sangue , Docetaxel , Relação Dose-Resposta a Droga , Feminino , Humanos , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/efeitos dos fármacos , Masculino , Camundongos , Neoplasias/sangue , Receptor ErbB-2/efeitos dos fármacos , Taxoides/uso terapêutico
4.
Clin Cancer Res ; 13(6): 1769-74, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17363531

RESUMO

PURPOSE: The primary objective was to establish the dose-limiting toxicity (DLT) and recommended phase II dose of 17-(allylamino)-17-demethoxygeldanamycin (17AAG) given twice a week. EXPERIMENTAL DESIGN: Escalating doses of 17AAG were given i.v. to cohorts of three to six patients. Dose levels for schedule A (twice weekly x 3 weeks, every 4 weeks) were 100, 125, 150, 175, and 200 mg/m(2) and for schedule B (twice weekly x 2 weeks, every 3 weeks) were 150, 200, and 250 mg/m(2). Peripheral blood mononuclear cells (PBMC) were collected for assessment of heat shock protein (HSP) 90 and HSP90 client proteins. RESULTS: Forty-four patients were enrolled, 32 on schedule A and 12 on schedule B. On schedule A at 200 mg/m(2), DLTs were seen in two of six patients (one grade 3 thrombocytopenia and one grade 3 abdominal pain). On schedule B, both patients treated at 250 mg/m(2) developed DLT (grade 3 headache with nausea/vomiting). Grade 3/4 toxicities seen in >5% of patients were reversible elevations of liver enzymes (47%), nausea (9%), vomiting (9%), and headache (5%). No objective tumor responses were observed. The only consistent change in PBMC proteins monitored was a 0.8- to 30-fold increase in HSP70 concentrations, but these were not dose dependent. The increase in PBMC HSP70 persisted throughout the entire cycle of treatment but returned to baseline between last 17AAG dose of cycle 1 and first 17AAG dose of cycle 2. CONCLUSIONS: The recommended phase II doses of 17AAG are 175 to 200 mg/m(2) when given twice a week and consistently cause elevations in PBMC HSP70.


Assuntos
Benzoquinonas/administração & dosagem , Benzoquinonas/farmacocinética , Lactamas Macrocíclicas/administração & dosagem , Lactamas Macrocíclicas/farmacocinética , Neoplasias/tratamento farmacológico , Adulto , Idoso , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Benzoquinonas/efeitos adversos , Células Sanguíneas/metabolismo , Relação Dose-Resposta a Droga , Feminino , Proteínas de Choque Térmico HSP70/sangue , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Lactamas Macrocíclicas/efeitos adversos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Neoplasias/sangue , Neoplasias/metabolismo , Neoplasias/patologia , Falha de Tratamento
5.
Clin Cancer Res ; 13(6): 1783-8, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17363533

RESUMO

PURPOSE: Heat shock protein 90 (Hsp90) is essential for the posttranslational control of many regulators of cell growth, differentiation, and apoptosis. 17-N-Allylamino-17-demethoxygeldanamycin (17-AAG) binds to Hsp90 and alters levels of proteins regulated by Hsp90. We conducted a phase I trial of 17-AAG in pediatric patients with recurrent or refractory neuroblastoma, Ewing's sarcoma, osteosarcoma, and desmoplastic small round cell tumor to determine the maximum tolerated dose, define toxicity and pharmacokinetic profiles, and generate data about molecular target modulation. EXPERIMENTAL DESIGN: Escalating doses of 17-AAG were administered i.v. over 1 to 2 h twice weekly for 2 weeks every 21 days until patients experienced disease progression or toxicity. harmacokinetic and pharmacodynamic studies were done during cycle 1. RESULTS: Fifteen patients were enrolled onto dose levels between 150 and 360 mg/m(2); 13 patients were evaluable for toxicity. The maximum tolerated dose was 270 mg/m(2). DLTs were grade 3 transaminitis and hypoxia. Two patients with osteosarcoma and bulky pulmonary metastases died during cycle 1 and were not evaluable for toxicity. No objective responses were observed. 17-AAG pharmacokinetics in pediatric patients were linear; clearance and half-life were 21.6 +/- 6.21 (mean +/- SD) L/h/m(2) and 2.6 +/- 0.95 h, respectively. Posttherapy increases in levels of the inducible isoform of Hsp70, a marker of target modulation, were detected in peripheral blood mononuclear cells at all dose levels. CONCLUSION: 17-AAG was well tolerated at a dose of 270 mg/m(2) administered twice weekly for 2 of 3 weeks. Caution should be used in treatment of patients with bulky pulmonary disease.


Assuntos
Benzoquinonas/administração & dosagem , Benzoquinonas/farmacocinética , Lactamas Macrocíclicas/administração & dosagem , Lactamas Macrocíclicas/farmacocinética , Neoplasias/tratamento farmacológico , Pediatria , Adolescente , Adulto , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Benzoquinonas/efeitos adversos , Biomarcadores Tumorais/análise , Criança , Pré-Escolar , Feminino , Humanos , Lactamas Macrocíclicas/efeitos adversos , Masculino , Dose Máxima Tolerável , Neoplasias/patologia , Recidiva , Falha de Tratamento
6.
Clin Cancer Res ; 12(19): 5817-25, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17020989

RESUMO

PURPOSE: Cisplatin adducts to nuclear DNA (nDNA) are felt to be the molecular lesions that trigger apoptosis, but the mechanism linking nDNA adduct formation and cell death is unclear. Some literature in the last decade has suggested a possible direct effect of cisplatin on mitochondria independent of nDNA interaction. In this study, we define separately the sequelae of cisplatin interactions with nDNA and with mitochondria in head and neck squamous cell carcinoma (HNSCC) cell lines. EXPERIMENTAL DESIGN: Cisplatin binding to mitochondrial DNA (mtDNA) and proteins was analyzed by atomic absorption spectroscopy and other methods. RESULTS: Following 1 hour of exposure to cisplatin, platinum adducts to mtDNA were 300- to 500-fold more abundant than adducts to nDNA; these differences were not due to differences in rates of adduct repair. Whereas HNSCC cell cytoplasts free of nDNA retained the same dose-dependent cisplatin sensitivity as parental cells, HNSCC rho(0) cells free of mtDNA were 4- to 5-fold more resistant to cisplatin than parental cells. Isolated mitochondria released cytochrome c within minutes of exposure to cisplatin, and ultrastructural analysis of intact HNSCC cells by electron microscopy showed marked mitochondrial disruption after 4 hours of cisplatin treatment, whereas the nucleus and other cellular structures remain intact. The very prompt release of cytochrome c from isolated mitochondria implies that apoptosis does not require alteration in mitochondrial gene transcription. Further, cisplatin binds preferentially to mitochondrial membrane proteins, particularly the voltage-dependent anion channel. CONCLUSIONS: Cisplatin binding to nDNA is not necessary for induction of apoptosis in HNSCC, which can result from direct action of cisplatin on mitochondria.


Assuntos
Apoptose , Cisplatino/metabolismo , DNA Mitocondrial/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Membranas Mitocondriais/metabolismo , Neoplasias de Células Escamosas/metabolismo , Canais de Ânion Dependentes de Voltagem/metabolismo , Antineoplásicos/metabolismo , Núcleo Celular/metabolismo , Citocromos c/metabolismo , Adutos de DNA , Reparo do DNA , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Neoplasias de Células Escamosas/tratamento farmacológico , Neoplasias de Células Escamosas/patologia , Células Tumorais Cultivadas
7.
Cancer Res ; 66(11): 5582-91, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16740693

RESUMO

Histone modifications and DNA methylation are epigenetic phenomena that play a critical role in many neoplastic processes, including silencing of tumor suppressor genes. One such histone modification, particularly at H3 and H4, is methylation at specific lysine (K) residues. Whereas histone methylation of H3-K9 has been linked to DNA methylation and aberrant gene silencing in cancer cells, no such studies of H3-K27 have been reported. Here, we generated a stable cell line overexpressing a dominant-negative point mutant, H3-K27R, to examine the role of that specific lysine in ovarian cancer. Expression of this construct resulted in loss of methylation at H3-K27, global reduction of DNA methylation, and increased expression of tumor suppressor genes. One of the affected genes, RASSF1, was shown to be a direct target of H3-K27 methylation-mediated silencing. By increasing DNA-platinum adduct formation, indicating increased access of the drug to target DNA sequences, removal of H3-K27 methylation resensitized drug-resistant ovarian cancer cells to the chemotherapeutic agent cisplatin. This increased platinum-DNA access was likely due to relaxation of condensed chromatin. Our results show that overexpression of mutant H3-K27 in mammalian cells represents a novel tool for studying epigenetic mechanisms and the Histone Code Hypothesis in human cancer. Such findings show the significance of H3-K27 methylation as a promising target for epigenetic-based cancer therapies.


Assuntos
Cisplatino/farmacologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Genes Supressores de Tumor , Histonas/genética , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Mutação Puntual , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Cromatina/metabolismo , Ilhas de CpG , Metilação de DNA , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Feminino , Inativação Gênica , Humanos , Lisina/genética , Proteínas Supressoras de Tumor/genética
8.
Clin Cancer Res ; 11(9): 3385-91, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15867239

RESUMO

PURPOSE: 17-(Allylamino)-17-demethoxygeldanamycin (17AAG), a benzoquinone antibiotic, down-regulates oncoproteins by binding specifically to heat shock protein 90 (HSP90). We did a phase I study of 17AAG to establish the dose-limiting toxicity and maximum tolerated dose and to characterize 17AAG pharmacokinetics and pharmacodynamics. EXPERIMENTAL DESIGN: Escalating doses of 17AAG were given i.v. over 1 or 2 hours on a weekly x 3 schedule every 4 weeks to cohorts of three to six patients. Plasma pharmacokinetics of 17AAG and 17-(amino)-17-demethoxygeldanamycin (17AG) were assessed by high-performance liquid chromatography. Expression of HSP70 and HSP90 in peripheral blood mononuclear cells was measured by Western blot. RESULTS: Forty-five patients were enrolled to 11 dose levels between 10 and 395 mg/m2. The maximum tolerated dose was 295 mg/m2. Dose-limiting toxicity occurred in both patients (grade 3 pancreatitis and grade 3 fatigue) treated with 395 mg/m2. Common drug-related toxicities (grade 1 and 2) were fatigue, anorexia, diarrhea, nausea, and vomiting. Reversible elevations of liver enzymes occurred in 29.5% of patients. Hematologic toxicity was minimal. No objective responses were observed. 17AAG pharmacokinetics was linear. Peak plasma concentration and area under the curve of 17AG, the active major metabolite of 17AAG, increased with 17AAG dose, but the relationships were more variable than with 17AAG. 17AAG and 17AG in plasma were >90% protein bound. There were no consistent changes in peripheral blood mononuclear cell HSP90 or HSP70 content. CONCLUSIONS: 17AAG doses between 10 and 295 mg/m2 are well tolerated. 17AAG pharmacokinetics is linear. Peripheral blood mononuclear cell HSP90 and HSP70 are uninformative pharmacodynamic markers. The dose recommended for future studies is 295 mg/m2 weekly x 3, repeated every 4 weeks.


Assuntos
Neoplasias/tratamento farmacológico , Rifabutina/análogos & derivados , Rifabutina/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Área Sob a Curva , Benzoquinonas , Diarreia/induzido quimicamente , Relação Dose-Resposta a Droga , Estradiol/sangue , Fadiga/induzido quimicamente , Feminino , Hormônio Foliculoestimulante/sangue , Proteínas de Choque Térmico HSP70/sangue , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/sangue , Humanos , Hidrocortisona/sangue , Lactamas Macrocíclicas , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Leucopenia/induzido quimicamente , Hormônio Luteinizante/sangue , Masculino , Pessoa de Meia-Idade , Náusea/induzido quimicamente , Neoplasias/sangue , Progesterona/sangue , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Rifabutina/efeitos adversos , Rifabutina/farmacocinética , Testosterona/sangue , Resultado do Tratamento , Vômito/induzido quimicamente
9.
Cancer Chemother Pharmacol ; 55(4): 333-342, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15723259

RESUMO

PURPOSE: Modulation of platinating agent cytotoxicity has important clinical implications as a result of their widespread use in the treatment of many different cancers. O6-Benzylguanine (BG) enhances the cytotoxicity of cisplatin against several human tumor lines. The purpose of our work was to elucidate whether BG affects pathways prior to DNA damage (i.e., glutathione, GSH) or following DNA damage (i.e., nucleotide excision repair, NER). METHODS: In efforts to determine the mechanism of enhancement we: (1) evaluated whether different sequences of BG plus cisplatin treatment differed in their ability to enhance cisplatin-induced cytotoxicity and DNA platination; (2) determined the effect of BG on GSH and glutathione S-transferase (GST) activity and; (3) determined whether BG enhanced cisplatin-induced cytotoxicity in cells lacking specific enzymes in the NER pathway. Colony-forming assay, atomic absorption spectroscopy and HPLC were employed to measure tumor cell growth inhibition, quantitate the amount of platinum on DNA, and determine intracellular GSH concentrations, respectively. RESULTS: Increased cytotoxicity and platination of DNA was observed when cells were exposed to BG prior to and/or during cisplatin treatment and not when BG followed cisplatin treatment. BG did not significantly alter GST activity with minimal depletion of GSH. In contrast, buthionine sulfoximine (BSO) caused a much more dramatic decrease in GSH than BG that was not accompanied by a dramatic increase in sensitivity to cisplatin. Furthermore, BG enhanced the cytotoxicity of cisplatin in a series of cell lines deficient in NER. CONCLUSIONS: Overall, our results suggest that the mechanism of enhancement involves neither the GSH nor the NER pathways, but triggers an event prior to DNA platination damage that ultimately results in increased cytotoxicity, apoptosis and increased platination levels.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Reparo do DNA/efeitos dos fármacos , Glutationa/metabolismo , Guanina/análogos & derivados , Guanina/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Dano ao DNA , Sinergismo Farmacológico , Humanos , Platina/metabolismo , Células Tumorais Cultivadas
10.
Cancer Chemother Pharmacol ; 55(3): 237-43, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15503027

RESUMO

PURPOSE: 17-(Allylamino)-17-demethoxygeldanamycin (17AAG) is a novel anticancer agent in clinical development. The objectives of this study were to develop a population pharmacokinetic model for 17AAG and its major metabolite, 17AG, and to investigate influences of patient characteristics and biochemical markers on pharmacokinetic parameters estimated for 17AAG and 17AG. EXPERIMENTAL DESIGN: In a phase I clinical study, 17AAG was administered by intravenous infusion to 43 patients with refractory, advanced malignancies. Plasma concentrations of 17AAG and 17AG were determined by high-performance liquid chromatography. Plasma concentration vs time data were modeled using NONMEM. Nine covariates (age, sex, performance status, weight, height, body surface area, AST, bilirubin and serum creatinine) were investigated for their influences on individual pharmacokinetic parameters. RESULTS: Plasma concentration vs time data were best described by a two-compartment model for 17AAG and a one-compartment model for 17AG. Volumes of distribution were 24.2 and 89.6 l for 17AAG. Total elimination clearances were 26.7 and 21.3 l/h for 17AAG and 17AG, respectively. Both fixed and random effects pharmacokinetic parameters were well estimated. None of the covariates explained the interindividual variability in 17AAG and 17AG pharmacokinetic parameters or improved the fit of the model based on objective function changes. CONCLUSIONS: A population pharmacokinetic model was developed to describe 17AAG and 17AG population pharmacokinetic parameters and interindividual variabilities. There were substantial interindividual variabilities in 17AAG and 17AG pharmacokinetic parameters despite BSA-normalized dosing.


Assuntos
Antineoplásicos/metabolismo , Neoplasias/metabolismo , Rifabutina/análogos & derivados , Rifabutina/farmacocinética , Adulto , Fatores Etários , Idoso , Benzoquinonas , Biomarcadores , Estatura , Peso Corporal , Feminino , Humanos , Lactamas Macrocíclicas , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Neoplasias/sangue , Fatores Sexuais
11.
J Pharmacol Exp Ther ; 312(1): 206-13, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15304523

RESUMO

Pharmacological inhibitors of cyclin-dependent kinase (CDK)2 are currently in preclinical and clinical development. The purpose of our work was to evaluate a series of guanine derivatives for their ability to inhibit CDK2, affect cell cycle progression, and enhance the cytotoxic and apoptotic effects of cisplatin. A panel of guanine derivatives, including O(6)-benzylguanine (O(6)-BG), S(6)-benzyl-6-thioguanine (S(6)-BG), S(6)-[(cyclohexyl)methyl]-6-thioguanine (S(6)-CMG), O(6)-[(cyclohexyl)methyl]guanine (O(6)-CMG), O(6)-benzyl-9-methylguanine (9-CH(3)-BG), O(6)-[(cyclohexyl)methyl]-9-methyl-guanine (9-CH(3)-CMG), and 7-benzylguanine (N7-BG), exhibited varying degrees of CDK2 inhibition with O(6)-CMG being the most potent and 9-CH(3)-BG, 9-CH(3)-CMG, and N7-BG the least potent compounds. Treatment with S(6)-CMG and O(6)-CMG significantly decreased the percentage of cells in S phase. In SQ20b and SCC61 head and neck cancer cell lines, the most potent CDK2 inhibitor, O(6)-CMG, was also the most effective at enhancing cisplatin-induced cytotoxicity and apoptosis. Cisplatin-induced DNA platination increased in SQ20b cells pretreated with S(6)-BG, S(6)-CMG, and O(6)-CMG. Treatment with both O(6)-BG and trichostatin A, an indirect cell cycle inhibitor, demonstrated additive effects on cisplatin-induced cytotoxicity. In summary, we have identified a group of guanine derivatives that were effective modulators of cisplatin-induced cytotoxicity and apoptosis.


Assuntos
Antineoplásicos/farmacologia , Quinases relacionadas a CDC2 e CDC28/antagonistas & inibidores , Ciclo Celular/efeitos dos fármacos , Cisplatino/farmacologia , Guanina/análogos & derivados , Guanina/farmacologia , Quinase 2 Dependente de Ciclina , DNA/efeitos dos fármacos , DNA/metabolismo , Combinação de Medicamentos , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Humanos , Ácidos Hidroxâmicos/farmacologia , O(6)-Metilguanina-DNA Metiltransferase/antagonistas & inibidores , Células Tumorais Cultivadas
12.
Anal Biochem ; 323(1): 19-25, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14622954

RESUMO

Although wortmannin is extensively used in molecular signaling studies, its stability in tissue culture medium has not been assessed precisely. Therefore, we used high-performance liquid chromatography (HPLC) and mass spectrometry (MS) to characterize the decomposition of wortmannin in five commonly used media. Wortmannin was added to medium alone or to medium supplemented with 10% unheated or heat-inactivated fetal bovine serum and incubated at 37 degrees C. After 0, 5, 10, 20, 35, and 60 min, wortmannin remaining in the medium was quantified, and its decay constant and half-life were calculated. In all media, wortmannin decomposed monoexponentially, with half-lives between 8 and 13 min. HPLC/MS indicated that wortmannin decomposed to materials with m/z 447, 433, 373, and 313. Acidification of material produced by incubation of wortmannin in tissue culture medium or 1 microM NaOH converted the material with m/z 447 back to one that cochromatographed with and had an m/z (429) identical to that of wortmannin. Therefore wortmannin is much less stable in tissue culture medium than previously thought although some apparent loss of wortmannin reflects reversible, pH-dependent opening of the lactone ring of wortmannin. This rapid and complex decomposition of wortmannin argues for care being taken in how it is used in in vitro studies.


Assuntos
Androstadienos/metabolismo , Cromatografia Líquida de Alta Pressão/métodos , Biodegradação Ambiental , Meios de Cultura/farmacologia , Estabilidade de Medicamentos , Meia-Vida , Espectrometria de Massas , Wortmanina
13.
Mol Cancer Ther ; 2(7): 633-40, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12883036

RESUMO

O(6)-Benzylguanine (O(6)-BG), a potent inactivator of the DNA repair protein O(6)-alkylguanine-DNA alkyltransferase (AGT), is presently in clinical trials combined with alkylating agents that modify the O(6) position of DNA guanine residues, i.e., 1,3-bis(2-chloroethyl)-1-nitrosourea and temozolomide. Previous work demonstrated that O(6)-BG also enhances the cytotoxicity of cyclophosphamide, ifosfamide, and nitrogen mustards in Chinese hamster ovary cells. We have extended this study to include other clinically relevant agents that form interstrand and intrastrand cross-links including cisplatin and carboplatin. Pretreatment of a series of head and neck tumor cell lines (i.e., SQ20b, JSQ3, SCC25, SCC35, and SCC61), Chinese hamster ovary cells, and HT29 human colon tumor cells with O(6)-BG (100 micro M for 2 h before treatment and 2 h during treatment) resulted in a 2-fold decrease in the ED(50) of cisplatin and a concomitant increase in the percentage of cells undergoing apoptosis. The enhancement was independent of AGT activity. Similar enhancement was observed with carboplatin, but no enhancement was seen in AGT-deficient cell lines with radiation or temozolomide, demonstrating the dependence of the effect on bifunctional, cross-linking agents. Furthermore, levels of platinum on DNA after treatment with cisplatin increased 1.4-fold in SQ20b cells and 4.5-fold in JSQ3 cells immediately after treatment with O(6)-BG plus cisplatin and remained elevated for 48 h. Consistent with greater cytotoxicity and apoptosis is the approximately 2-fold higher amount of DNA damage when cells are treated with O(6)-BG plus cisplatin compared with cisplatin alone. Modulation of cisplatin therapy with O(6)-BG might improve the prognosis of patients with head and neck, ovarian, testicular, or lung cancer who are treated with this drug.


Assuntos
Apoptose/efeitos dos fármacos , Carboplatina/farmacologia , Cisplatino/toxicidade , Dano ao DNA , DNA de Neoplasias/efeitos dos fármacos , Guanina/análogos & derivados , Guanina/farmacologia , Animais , Células CHO/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Cricetinae , Interações Medicamentosas , Sinergismo Farmacológico , Humanos , O(6)-Metilguanina-DNA Metiltransferase/antagonistas & inibidores , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Células Tumorais Cultivadas
14.
Cancer Chemother Pharmacol ; 52(2): 139-46, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12761648

RESUMO

PURPOSE: 17-(Allylamino)-17-demethoxygeldanamycin (17AAG), an analogue of the benzoquinone ansamycin geldanamycin, has been extensively studied preclinically and is being evaluated clinically. Studies were performed to define the biliary excretion of 17AAG after i.v. delivery to rats, and to characterize the metabolites of 17AAG observed in rat bile. MATERIALS AND METHODS: In vivo studies were performed in bile-duct-cannulated Fischer 344 rats given a 10 mg/kg i.v. bolus dose of 17AAG. In vitro studies were performed with cloned human CYPs and microsomal epoxide hydrolase. Biliary excretion of 17AAG and metabolites was quantified by HPLC and followed for 4 h after drug delivery. 17AAG metabolites in bile and in in vitro reaction mixtures were identified with LC/MS/MS. RESULTS: By 15 min after i.v. delivery of 17AAG, bile contained at least 15 biotransformation products with absorbance spectra similar to that of 17AAG. Of these, metabolites eluting at 2.7, 2.9, and 8.6 min were present in sufficient concentrations to be quantified, although the lack of authentic standards resulted in their being expressed as 17AAG equivalents. Within the first 4 h after drug delivery, biliary excretion accounted for 28.9+/-6.1% of the 10-mg/kg 17AAG dose. 17AAG and 17-(amino)-17-demethoxygeldanamycin (17AG) accounted for 4.1+/-1.0% of the delivered dose, with 17AAG accounting for 2.0+/-0.5% and 17AG accounting for 2.1+/-0.5%. The metabolites eluting at 2.7, 2.9, and 8.6 min accounted for 10.6+/-2.0%, 9.8+/-1.2%, and 1.0+/-0.2%, respectively, of the administered dose. LC/MS/MS analysis of bile demonstrated major metabolites with molecular weights of 545 and 619, corresponding to 17AG and the diol previously described as resulting from metabolism of 17AAG by CYP3A and microsomal epoxide hydrolase. Of the remaining proposed metabolites, ten had a mass and MS/MS spectrum consistent with mono-oxygenated 17AAG metabolites. One of these metabolites has been identified as the epoxide previously described as resulting from CYP3A oxidation of the allyl double bond. Two other proposed metabolites had a mass and MS/MS spectrum consistent with demethylated 17AAG metabolites, and one had a mass and MS/MS spectrum consistent with a di-demethylated 17AAG metabolite. An analogous series of demethylated and oxidized metabolites was also observed for the 17AG metabolite. CONCLUSIONS: Biliary excretion of 17AAG represents a major route of elimination, although most of the material excreted is in the form of metabolites. Bile of rats dosed with 17AAG contained a number of metabolites not previously identified in the plasma or urine of mice treated with 17AAG, but analogous to metabolites described in bile of rats treated with 17-(dimethylaminoethylamino)-17-demethoxygeldanamycin (17DMAG, NSC 707545), another geldanamycin analogue undergoing preclinical evaluation in preparation for subsequent clinical trials.


Assuntos
Antibióticos Antineoplásicos/metabolismo , Bile/metabolismo , Rifabutina/análogos & derivados , Rifabutina/metabolismo , Animais , Antibióticos Antineoplásicos/química , Benzoquinonas , Biotransformação , Cromatografia Líquida de Alta Pressão , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/química , Epóxido Hidrolases/química , Humanos , Lactamas Macrocíclicas , Masculino , Espectrometria de Massas , Oxirredução , Ratos , Ratos Endogâmicos F344 , Rifabutina/química
15.
J Clin Oncol ; 21(4): 697-703, 2003 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-12586808

RESUMO

PURPOSE: To determine maximum-tolerated dose, toxicities, and pharmacokinetics associated with weekly intravesical gemcitabine therapy in patients with superficial bladder cancer. PATIENTS AND METHODS: Fifteen patients with recurrent superficial transitional cell bladder carcinoma who experienced prior intravesical therapy failure were studied. Two to 4 weeks after complete transurethral resection, gemcitabine was administered intravesically, once weekly for 6 consecutive weeks. Dwell time was 2 hours. Pharmacokinetics of gemcitabine and its metabolite, 2'2'-difluorodeoxyuridine (dFdU), were studied in plasma and urine. Cystoscopy was repeated 6 weeks after therapy. RESULTS: Three-patient cohorts were enrolled sequentially at doses of 500, 1,000, and 1,500 mg in 100 mL 0.9% NaCl. Two patients received 2,000 mg in 100 mL. An additional four patients received 2,000 mg in 50 mL. No grade 4 toxicity or clinically relevant myelosuppression was noted. Nine of 13 evaluable patients were recurrence-free at 12 weeks. Low concentrations of gemcitabine (< or = 1 microg/mL) were present transiently in plasma of all patients receiving 2,000 mg in 50 mL. Gemcitabine was undetectable in plasma of other patients. dFdU was undetectable in plasma of patients receiving less than 1,500 mg. At doses > or = 1,500 mg, dFdU concentrations increased until 90 to 120 minutes and then declined little, if any. Plasma dFdU concentrations implied absorption of 0.5% to 5.5% of instilled dose. Between 61% and 100% of the gemcitabine dose was accounted for in voided urine. No dFdU was measured in voided urine. CONCLUSION: Intravesical gemcitabine, at doses up to 2 g/wk, is well tolerated, is associated with minimal systemic absorption, and has promising efficacy in treatment of superficial bladder cancer.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Carcinoma de Células de Transição/tratamento farmacológico , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapêutico , Neoplasias da Bexiga Urinária/tratamento farmacológico , Administração Intravesical , Idoso , Idoso de 80 Anos ou mais , Antimetabólitos Antineoplásicos/sangue , Antimetabólitos Antineoplásicos/farmacocinética , Carcinoma de Células de Transição/cirurgia , Cromatografia Líquida de Alta Pressão , Desoxicitidina/sangue , Desoxicitidina/farmacocinética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias da Bexiga Urinária/cirurgia , Gencitabina
16.
Cancer Res ; 63(2): 312-8, 2003 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-12543781

RESUMO

Cisplatin is a potent cytotoxic agent that functions as a bivalent electrophile, forming both interstrand and intrastrand DNA cross-links. Cisplatin-mediated DNA damage results in cell cycle arrest and initiation of apoptotic cell death. Increased cellular glutathione concentrations have been closely correlated with cisplatin resistance but do not reduce the extent of cisplatin-DNA adduct formation. One hypothesis to explain the ability of glutathione to inhibit cisplatin cytotoxicity is that glutathione, through its antioxidant function, plays a role in apoptotic regulatory pathways. We tested this hypothesis using MCF-7 breast cancer cells transfected with the apoptotic inhibitor Bcl-2. Bcl-2 overexpression in MCF-7 cells was associated with a nearly 3-fold increase in cellular glutathione levels and with increased resistance to cell death after cisplatin exposure. Treatment of MCF-7 lines with buthionine sulfoximine, an inhibitor of glutathione synthesis, normalized glutathione levels in Bcl-2 and control transfectants and completely abrogated Bcl-2-mediated cisplatin resistance without affecting Bcl-2 expression. Bcl-2 overexpression and up-regulation of glutathione were not associated with a change in either cisplatin-DNA adduct formation or repair over time. These results suggest that Bcl-2-mediated cisplatin resistance in MCF-7 cells is dependent on up-regulation of glutathione production, which contributes to cell survival by mechanisms independent of cisplatin inactivation or inhibition of DNA adduct formation. A similar dependence on glutathione for Bcl-2-mediated inhibition of cisplatin toxicity was confirmed in a second cell line, the lymphocytic precursor FL5.12. Taken together, these data suggest that apoptotic signaling after genotoxic exposure can be inhibited by the antioxidant activity of glutathione. Inhibition of glutathione synthesis or modulation of glutathione stores in tumors that overexpress Bcl-2 may comprise a novel anticancer strategy.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Glutationa/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Dano ao DNA , DNA de Neoplasias/metabolismo , Resistencia a Medicamentos Antineoplásicos , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glutamato-Cisteína Ligase/biossíntese , Glutamato-Cisteína Ligase/genética , Glutamato-Cisteína Ligase/metabolismo , Glutationa/biossíntese , Glutationa Sintase/biossíntese , Glutationa Sintase/metabolismo , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/genética , Transfecção , Células Tumorais Cultivadas , Regulação para Cima , gama-Glutamiltransferase/biossíntese , gama-Glutamiltransferase/genética , gama-Glutamiltransferase/metabolismo
17.
Mol Cancer Ther ; 1(10): 821-9, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12492115

RESUMO

1,25-Dihydroxyvitamin D3 (1,25D3) exhibits potent antitumor activity in the murine squamous cell carcinoma (SCC) SCCVII/SF, and the combination of 1,25D3 with cisplatin (1,25D3/cisplatin) demonstrates even greater activity. Because these agents possess different mechanisms of cytotoxicity, studies were initiated to define the mechanism by which the combination displays enhanced activity. Median dose-effect analysis demonstrates that 1,25D3 and cisplatin act synergistically to inhibit SCC growth. When SCC cells were treated with 1,25D3 (10 nM) and/or cisplatin (0.5 microg/ml), greater caspase-3 activation was observed for the combination than for either agent alone. This suggests that the enhanced cytotoxicity is, at least in part, due to greater induction of apoptosis. No alterations in cellular platinum concentration or platinum-DNA adducts were observed for 1,25D3/cisplatin cotreatment compared with cisplatin treatment alone. Effects of the combination on cisplatin and 1,25D3 signaling pathways in adherent (nonapoptotic) and floating (apoptotic) cells were explored. Cisplatin induced p53 and its downstream targets, p21(Cip1) (p21) and Bax, in both cell populations. In contrast, 1,25D3 reduced p53, p21, and Bax to nearly undetectable levels in adherent cells. In the floating cells, 1,25D3 reduced levels of p53 and p21, but Bax expression was maintained at control levels. Expression of these proteins in cells treated with 1,25D3/cisplatin was similar to treatment with 1,25D3 alone. The two agents also had divergent effects on survival and stress signaling pathways. Phospho-extracellular signal-regulated kinase 1/2 and phospho-Jun levels increased after treatment with cisplatin but decreased after treatment with 1,25D3 and 1,25D3/cisplatin. Moreover, cisplatin decreased levels of mitogen-activated protein kinase kinase kinase (MEKK-1), whereas 1,25D3 up-regulated MEKK-1, and 1,25D3/cisplatin further up-regulated MEKK-1. We propose that the increased cytotoxicity for 1,25D3/cisplatin results from cisplatin enhancement of 1,25D3-induced apoptotic signaling through MEKK-1.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Calcitriol/farmacologia , Cisplatino/farmacologia , Regulação Neoplásica da Expressão Gênica , MAP Quinase Quinase Quinase 1 , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Serina-Treonina Quinases/biossíntese , Transdução de Sinais , Animais , Western Blotting , Sobrevivência Celular , Corantes/farmacologia , Adutos de DNA , Relação Dose-Resposta a Droga , Camundongos , Proteína Quinase 3 Ativada por Mitógeno , Modelos Biológicos , Sais de Tetrazólio/farmacologia , Tiazóis/farmacologia , Células Tumorais Cultivadas , Regulação para Cima
18.
Clin Cancer Res ; 8(9): 2992-9, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12231546

RESUMO

One possible explanation for variable tumor response within a single patient may be related to delivery of chemotherapeutic agents to the tumors. Microdialysis was used to evaluate inter- and intratumoral disposition of unbound platinum (Pt) after cisplatin administration to mice bearing B16 murine melanoma tumors or H23 human NSCLC xenografts. Before i.v. dosing with cisplatin (3 or 10 mg/kg), microdialysis probes were placed into the right and left sides of each tumor, and serial extracellular fluid (ECF) samples were collected for 2 h. After microdialysis, tumor samples were obtained at each probe site to measure total Pt and Pt-DNA adducts. In a separate study, serial plasma samples (n = 3 mice/time point) were obtained between 5 min and 2 h. Unbound Pt in tumor ECF and plasma and total Pt in tumor homogenates were measured by flameless atomic absorption spectrophotometry. Pt-DNA adducts in tumor samples were measured via (32)P-postlabeling. Area under the plasma (AUC(P)) and tumor ECF (AUC(ECF)) concentration-time curves of unbound Pt were calculated. Factor VIII expression was measured by immunohistochemistry in tumor samples. After administration of 3 or 10 mg/kg of cisplatin to mice bearing B16 tumors, there was a proportional increase in AUC(PL) with dose; however, there was not a proportional increase in AUC(ECF). There was a relatively high (30-fold) inter- and low (2.5-fold) intratumoral variability in AUC(ECF). AUC(ECF) correlated better with Pt-DNA adduct formation than did total Pt concentration in tumors. There was no relationship between Factor VIII expression and Pt exposure in tumors. The variable penetration of Pt from plasma into tumor ECF may be associated with variable response of tumors.


Assuntos
Antineoplásicos/farmacocinética , Carcinoma Pulmonar de Células não Pequenas/química , Cisplatino/farmacocinética , Neoplasias Pulmonares/química , Melanoma Experimental/química , Platina/análise , Animais , Antineoplásicos/administração & dosagem , Área Sob a Curva , Carcinoma Pulmonar de Células não Pequenas/irrigação sanguínea , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Cisplatino/administração & dosagem , Adutos de DNA , DNA de Neoplasias/efeitos dos fármacos , Fator VIII/biossíntese , Fator VIII/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Microdiálise , Modelos Biológicos , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Neovascularização Patológica/tratamento farmacológico , Organismos Livres de Patógenos Específicos , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Clin Cancer Res ; 8(7): 2437-42, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12114450

RESUMO

PURPOSE: Gemcitabine is an excellent candidate for regional therapy. We quantified cerebrospinal fluid (CSF) and plasma concentrations of gemcitabine and its inactive metabolite, 2',2'-difluorodeoxyuridine (dFdU), in nonhuman primates given intrathecal gemcitabine. EXPERIMENTAL DESIGN: Three nonhuman primates received 5 mg of gemcitabine via lateral ventricle. CSF was sampled from the fourth ventricle in all of the animals and the lumbar space in one, and one had plasma sampled. One animal had ventricular CSF sampled after receiving 5 mg intralumbar gemcitabine. Gemcitabine and dFdU were measured by high-performance liquid chromatography. Three additional animals had 5 mg intralumbar gemcitabine administered weekly for 4 weeks and were monitored for toxicity. RESULTS: At 37 degrees C in vitro, gemcitabine was stable in CSF. Ventricular delivery of gemcitabine produced peak ventricular CSF gemcitabine concentrations of 297 +/- 105 microg/ml. After 6 h, the concentrations were <0.03 microg/ml. Intrathecal gemcitabine was rapidly and extensively converted to dFdU. CSF dFdU concentrations increased to 82 microg/ml at 1 h and then declined to very low values by 24 h. After intraventricular administration, CSF gemcitabine and dFdU area(s) under the curve (AUC) were 251 +/- 85 and 249 +/- 88 microg/ml x h. Intralumbar gemcitabine produced lower ventricular CSF gemcitabine and dFdU concentrations than did intraventricular gemcitabine. The plasma gemcitabine AUC associated with 5 mg of intraventricular gemcitabine was 2 mg/ml x h, which was >200-fold lower than the CSF gemcitabine AUC in the same animal. Transient CSF pleocytosis was the only toxicity observed. CONCLUSIONS: Our results demonstrate a large pharmacokinetic advantage of intrathecal gemcitabine and support a planned Phase I clinical trial of this dosing strategy.


Assuntos
Antimetabólitos Antineoplásicos/farmacocinética , Líquido Cefalorraquidiano/metabolismo , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacocinética , Animais , Área Sob a Curva , Cromatografia Líquida de Alta Pressão , Injeções Intraventriculares , Macaca mulatta , Masculino , Taxa de Depuração Metabólica , Gencitabina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA