Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Lancet Oncol ; 13(8): 773-81, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22805291

RESUMEN

BACKGROUND: Inhibition of MEK stops cell proliferation and induces apoptosis; therefore, this enzyme is a key anticancer target. Trametinib is a selective, orally administered MEK1/MEK2 inhibitor. We aimed to define the maximum tolerated dose and recommended phase 2 dose of trametinib and to assess its safety, pharmacokinetics, pharmacodynamics, and response rate in individuals with advanced solid tumours. METHODS: We undertook a multicentre phase 1 study in patients with advanced solid tumours and adequate organ function. The study was in three parts: dose escalation to define the maximum tolerated dose; identification of the recommended phase 2 dose; and assessment of pharmacodynamic changes. Intermittent and continuous dosing regimens were analysed. Blood samples and tumour biopsy specimens were taken to assess pharmacokinetic and pharmacodynamic changes. Adverse events were defined with common toxicity criteria, and tumour response was measured by Response Evaluation Criteria In Solid Tumors. This study is registered with ClinicalTrials.gov, number NCT00687622. FINDINGS: We enrolled 206 patients (median age 58·5 years, range 19-92). Dose-limiting toxic effects included rash (n=2), diarrhoea (n=1), and central serous retinopathy (n=2). The most common treatment-related adverse events were rash or dermatitis acneiform (n=165; 80%) and diarrhoea (87; 42%), most of which were grade 1 and 2. The maximum tolerated dose was 3 mg once daily and the recommended phase 2 dose was 2 mg a day. The effective half-life of trametinib was about 4 days. At the recommended phase 2 dose, the exposure profile of the drug showed low interpatient variability and a small peak:trough ratio of 1·81. Furthermore, mean concentrations in plasma were greater than the preclinical target concentration throughout the dosing interval. Pathway inhibition and clinical activity were seen, with 21 (10%) objective responses recorded. INTERPRETATION: The recommended phase 2 dose of 2 mg trametinib once a day is tolerable, with manageable side-effects. Trametinib's inhibition of the expected target and clinical activity warrants its further development as a monotherapy and in combination. FUNDING: GlaxoSmithKline.


Asunto(s)
Antineoplásicos/administración & dosificación , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 2/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/administración & dosificación , Piridonas/administración & dosificación , Pirimidinonas/administración & dosificación , Administración Oral , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Área Bajo la Curva , Biopsia , Esquema de Medicación , Monitoreo de Drogas , Femenino , Semivida , Humanos , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Masculino , Dosis Máxima Tolerada , Tasa de Depuración Metabólica , Persona de Mediana Edad , Terapia Molecular Dirigida , Neoplasias/enzimología , Neoplasias/patología , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/farmacocinética , Piridonas/efectos adversos , Piridonas/farmacocinética , Pirimidinonas/efectos adversos , Pirimidinonas/farmacocinética , Resultado del Tratamiento , Estados Unidos , Adulto Joven
2.
Invest New Drugs ; 29(5): 1057-65, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20306339

RESUMEN

PURPOSE: The primary aim of this study was to characterize the 6-month overall survival and toxicity associated with second-line capecitabine treatment of advanced pancreatic cancer patients harboring the TYMS *2/*2 allele. The secondary aim was to analyze the response rate and pharmacokinetics of capecitabine-based therapy in this patient population. Lastly, TYMS, ATM and RecQ1 single nucleotide polymorphism were analyzed relative to overall survival in patients screened for study participation. METHODS: Eighty patients with stage IV pancreatic cancer were screened for the *2/*2 TYMS allele. Patients with the *2/*2 TYMS polymorphism were treated with capecitabine, 1,000 mg/m2 twice daily for 14 consecutive days of a 21 day cycle. Screened patients not possessing TYMS *2/*2 were monitored for survival. Pharmacokinetic analysis was done during Cycle 1 of the therapy. RESULTS: Sixteen of the 80 screened patients tested positive for *2/*2 TYMS variant. Four out of the 16 eligible patients were treated on study. The study was terminated early due to poor accrual and increased toxicity. Three patients experienced grade 3 non-hematologic toxicities of palmer-plantar erythrodysesthesia, diarrhea, nausea and vomiting. Grade 2 toxicities were similar and occurred in all patients. Only one patient was evaluable for response after completion of three cycles of therapy. The presence of the *2/*2 TYMS genotype in all of the screened patients trended toward a decreased overall survival. CONCLUSION: To our knowledge, this study represents the first genotype-directed clinical trial for patients with pancreatic adenocarcinoma. Although the study was closed early, it appears capecitabine therapy in pancreatic cancer patients harboring the TYMS *2/*2 variant may be associated with increased non-hematologic toxicity. This study also demonstrates the challenges performing a genotype-directed study in the second-line setting for patients with advanced pancreatic cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Desoxicitidina/análogos & derivados , Elementos de Facilitación Genéticos/genética , Fluorouracilo/análogos & derivados , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Timidilato Sintasa/genética , Administración Oral , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Capecitabina , Demografía , Desoxicitidina/administración & dosificación , Desoxicitidina/efectos adversos , Desoxicitidina/uso terapéutico , Femenino , Fluorouracilo/administración & dosificación , Fluorouracilo/efectos adversos , Fluorouracilo/uso terapéutico , Genotipo , Homocigoto , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/genética , Polimorfismo de Nucleótido Simple/genética , Análisis de Supervivencia , Resultado del Tratamiento , Gemcitabina
3.
Invest New Drugs ; 29(2): 340-6, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20084425

RESUMEN

BACKGROUND: 2-methoxyestradiol (2ME2) is an estradiol-17ß metabolite with antiproliferative and antiangiogenic activities. ENMD-1198 is an analog of 2ME2 which was developed to decrease the metabolism and increase both the bioavailability and antitumor activities of the parent molecule. This first-in-human phase I study evaluated the tolerability, pharmacokinetics and preliminary evidence of activity of ENMD-1198 in advanced cancer patients. METHODS: Eligible patients received ENMD-1198 orally once daily in Part A (standard 3 + 3 dose escalation design), or in Part B (accelerated dose escalation design). Cycle 1 consisted of 28 days daily dosing followed by a 14-(Part A) or 7-(Part B) day observation period, then continuously in 28 day cycles thereafter. RESULTS: A total of 29 patients were enrolled in 12 dose cohorts (5 to 550 mg/m²)/d). The most common drug-related toxicities were Grade 1/2 fatigue (55%), nausea and vomiting (37%), and constipation (34%). Two DLTs (Grade 4 neutropenia) occurred at 550 mg/m²/day, and 425 mg/m²/d was declared the maximum tolerated dose. ENMD-1198 was absorbed rapidly with a T(max) of 1-2 h. Exposure to ENMD-1198 (C(max) and AUC0₋24 hr increased linearly with dose. The mean terminal half-life was 15 h. A 3-fold accumulation was found after multiple doses. Five patients achieved stabilization of disease for at least 2 cycles, three of whom (with neuroendocrine carcinoma of pancreas, prostate cancer and ovarian cancer) demonstrated prolonged stabilization ranging from 8-24.5 cycles. CONCLUSION: ENMD-1198 is well-tolerated with a pharmacokinetic exposure profile compatible with once daily dosing. The recommended phase II dose of ENMD-1198 is 425 mg/m²/d. Early evidence of prolonged disease stabilization in pre-treated patients suggests ENMD-1198 is worthy of additional investigation.


Asunto(s)
Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Estradiol/análogos & derivados , Estrenos/farmacocinética , Estrenos/uso terapéutico , Neoplasias/tratamiento farmacológico , 2-Metoxiestradiol , Administración Oral , Adulto , Anciano , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Demografía , Perros , Relación Dosis-Respuesta a Droga , Estradiol/administración & dosificación , Estradiol/efectos adversos , Estradiol/farmacocinética , Estradiol/uso terapéutico , Estrenos/administración & dosificación , Estrenos/efectos adversos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/sangre , Ratas , Especificidad de la Especie
4.
Anticancer Res ; 25(4): 3055-60, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16080565

RESUMEN

INTRODUCTION: Esophageal adenocarcinoma (EC) is increasing in incidence. Chemoradiation (CRT) is regarded as an acceptable alternative to surgery for the management of locally advanced EC. Ten-20% of EC patients are over the age of 75 years. There are limited data regarding efficacy and tolerability of CRT for the treatment of EC in the elderly. PATIENTS AND METHODS: We retrospectively reviewed EC cases > or = 70 years of age treated with CRT at a single institution. Clinical data, regarding therapy administered and outcome, were obtained from records. Clinical prognostic variables were analyzed against survival in a univariate model using the log rank test and in a multivariate model using Cox proportional hazards analysis. RESULTS: Thirty consecutive patient records were identified. Commonly used chemotherapy agents included 5-fluorouracil, cisplatin, paclitaxel and oxaliplatin. There was no significant correlation between age and survival. The dose of chemotherapy or radiation was unrelated to any of the toxicities (p-values > 0.16). The most common grade 3 or 4 toxicities were dehydration, hypotension, mucositis and pneumonitis. On multivariate analysis, adenocarcinoma histology (p = 0.0094) and higher radiation dose (p = 0.0158) were associated with improved survival. The median survival of the patients was 10 months. CONCLUSION: CRT was tolerable for older patients with EC. Close monitoring for dehydration, nutritional compromise and pulmonary toxicity is required.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/radioterapia , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/radioterapia , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/radioterapia , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Cisplatino/administración & dosificación , Terapia Combinada , Femenino , Fluorouracilo/administración & dosificación , Fluorouracilo/uso terapéutico , Humanos , Masculino , Compuestos Organoplatinos/administración & dosificación , Oxaliplatino , Paclitaxel/uso terapéutico , Estudios Retrospectivos , Tasa de Supervivencia
5.
Cancer Med ; 1(2): 207-17, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23342270

RESUMEN

Src tyrosine kinases are overexpressed in pancreatic cancers, and the oral Src inhibitor saracatinib has shown antitumor activity in preclinical models of pancreas cancer. We performed a CTEP-sponsored Phase II clinical trial of saracatinib in previously treated pancreas cancer patients, with a primary endpoint of 6-month survival. A Simon MinMax two-stage phase II design was used. Saracatinib (175 mg/day) was administered orally continuously in 28-day cycles. In the unselected portion of the study, 18 patients were evaluable. Only two (11%) patients survived for at least 6 months, and three 6-month survivors were required to move to second stage of study as originally designed. The study was amended as a biomarker-driven trial (leucine rich repeat containing protein 19 [LRRC19] > insulin-like growth factor-binding protein 2 [IGFBP2] "top scoring pairs" polymerase chain reaction [PCR] assay, and PIK3CA mutant) based on preclinical data in a human pancreas tumor explant model. In the biomarker study, archival tumor tissue or fresh tumor biopsies were tested. Biomarker-positive patients were eligible for the study. Only one patient was PIK3CA mutant in a 3' untranslated region (UTR) portion of the gene. This patient was enrolled in the study and failed to meet the 6-month survival endpoint. As the frequency of biomarker-positive patients was very low (<3%), the study was closed. Although we were unable to conclude whether enriching for a subset of second/third line pancreatic cancer patients treated with a Src inhibitor based on a biomarker would improve 6-month survival, we demonstrate that testing pancreatic tumor samples for a biomarker-driven, multicenter study in metastatic pancreas cancer is feasible.


Asunto(s)
Antineoplásicos/administración & dosificación , Benzodioxoles/administración & dosificación , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Quinazolinas/administración & dosificación , Familia-src Quinasas/antagonistas & inhibidores , Regiones no Traducidas 3'/genética , Administración Oral , Adulto , Anciano , Antineoplásicos/efectos adversos , Benzodioxoles/efectos adversos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Fosfatidilinositol 3-Quinasa Clase I , Femenino , Humanos , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Neoplasias Pancreáticas/mortalidad , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Quinazolinas/efectos adversos , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo
6.
Clin Cancer Res ; 18(4): 1051-62, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22173548

RESUMEN

PURPOSE: Despite the availability of several active combination regimens for advanced colorectal cancer (CRC), the 5-year survival rate remains poor at less than 10%, supporting the development of novel therapeutic approaches. In this study, we focused on the preclinical assessment of a rationally based combination against KRAS-mutated CRC by testing the combination of the MEK inhibitor, selumetinib, and vorinostat, a histone deacetylase (HDAC) inhibitor. EXPERIMENTAL DESIGN: Transcriptional profiling and gene set enrichment analysis (baseline and posttreatment) of CRC cell lines provided the rationale for the combination. The activity of selumetinib and vorinostat against the KRAS-mutant SW620 and SW480 CRC cell lines was studied in vitro and in vivo. The effects of this combination on tumor phenotype were assessed using monolayer and 3-dimensional cultures, flow cytometry, apoptosis, and cell migration. In vivo, tumor growth inhibition, (18)F-fluoro-deoxy-glucose positron emission tomography (FDG-PET), and proton nuclear magnetic resonance were carried out to evaluate the growth inhibitory and metabolic responses, respectively, in CRC xenografts. RESULTS: In vitro, treatment with selumetinib and vorinostat resulted in a synergistic inhibition of proliferation and spheroid formation in both CRC cell lines. This inhibition was associated with an increase in apoptosis, cell-cycle arrest in G(1), and reduced cellular migration and VEGF-A secretion. In vivo, the combination resulted in additive tumor growth inhibition. The metabolic response to selumetinib and vorinostat consisted of significant inhibition of membrane phospholipids; no significant changes in glucose uptake or metabolism were observed in any of the treatment groups. CONCLUSION: These data indicate that the rationally based combination of the mitogen-activated protein kinase/extracellular signal-regulated kinase inhibitor, selumetinib, with the HDAC inhibitor vorinostat results in synergistic antiproliferative activity against KRAS-mutant CRC cell lines in vitro. In vivo, the combination showed additive effects that were associated with metabolic changes in phospholipid turnover, but not on FDG-PET, indicating that the former is a more sensitive endpoint of the combination effects.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Bencimidazoles/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Mutación , Proteínas Proto-Oncogénicas/genética , Proteínas ras/genética , Animales , Apoptosis/efectos de los fármacos , Bencimidazoles/administración & dosificación , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Análisis por Conglomerados , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Femenino , Perfilación de la Expresión Génica , Inhibidores de Histona Desacetilasas/administración & dosificación , Humanos , Ácidos Hidroxámicos/administración & dosificación , Ligandos , Ratones , Ratones Desnudos , Resonancia Magnética Nuclear Biomolecular , Tomografía de Emisión de Positrones , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas p21(ras) , Transducción de Señal/efectos de los fármacos , Vorinostat , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas ras/metabolismo
7.
Mol Cancer Ther ; 9(12): 3351-62, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20923857

RESUMEN

Mutant K-ras activity leads to the activation of the RAS/RAF/MEK/ERK pathway in approximately 44% of colorectal cancer (CRC) tumors. Accordingly, several inhibitors of the MEK pathway are under clinical evaluation in several malignancies including CRC. The aim of this study was to develop and characterize predictive biomarkers of response to the MEK1/2 inhibitor AZD6244 in CRC in order to maximize the clinical utility of this agent. Twenty-seven human CRC cell lines were exposed to AZD6244 and classified according to the IC(50) value as sensitive (≤ 0.1 µmol/L) or resistant (>1 µmol/L). All cell lines were subjected to immunoblotting for effector proteins, K-ras/BRAF mutation status, and baseline gene array analysis. Further testing was done in cell line xenografts and K-ras mutant CRC human explants models to develop a predictive genomic classifier for AZD6244. The most sensitive and resistant cell lines were subjected to differential gene array and pathway analyses. Members of the Wnt signaling pathway were highly overexpressed in cell lines resistant to AZD6244 and seem to be functionally involved in mediating resistance by shRNA knockdown studies. Baseline gene array data from CRC cell lines and xenografts were used to develop a k-top scoring pair (k-TSP) classifier, which predicted with 71% accuracy which of a test set of patient-derived K-ras mutant CRC explants would respond to AZD6244, providing the basis for a patient-selective clinical trial. These results also indicate that resistance to AZD6244 may be mediated, in part, by the upregulation of the Wnt pathway, suggesting potential rational combination partners for AZD6244 in CRC.


Asunto(s)
Bencimidazoles/farmacología , Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Mutación/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas/genética , Proteínas ras/genética , Algoritmos , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Immunoblotting , Ratones , Ratones Desnudos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Proteínas Proto-Oncogénicas p21(ras) , Reproducibilidad de los Resultados , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Urol ; 174(2): 753-6, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16006970

RESUMEN

PURPOSE: : Interleukin-6 (IL-6) has an important role during prostate cancer progression and IL-6 levels in the serum of patients with hormone refractory and metastatic prostate cancer are significantly increased compared with those in patients with hormone sensitive and localized prostate cancer. The G>C polymorphism at position -174 in the promoter of the IL-6 gene has been associated with differences in IL-6 transcription in vitro and IL-6 protein levels in vivo. We determined the association of IL-6 polymorphism with prostate cancer progression. MATERIALS AND METHODS: : We examined the association of IL-6 polymorphism with the risk of advanced disease in 95 patients with different stages of prostate cancer using the tetra-primer polymerase chain reaction genotyping method. RESULTS: : We found that the -174G>C genotype of IL-6 gene was associated with an overall increased risk of advanced prostate cancer. A strong association between this genotype and Gleason score was observed at the -174G>C locus of the IL-6 gene (p <0.001). The distribution of this genotype was also significantly different between stages T3-T4 and T1-T2 tumors (p <0.001). In addition, the IL-6 genotype was linked with vascular invasion (p = 0.024), seminal vesicle involvement (p = 0.006) and capsular invasion (p <0.001). Furthermore, the -174G>C genotype of the IL-6 gene was significantly associated with increased serum prostate specific antigen (p = 0.004) and with recurrent prostate cancer compared with GG homozygotes (p = 0.027). CONCLUSIONS: : These data demonstrate a strong association of the -174G>C polymorphism of the IL-6 gene with the aggressiveness and recurrence of prostate cancer, suggesting that genetic predisposition of genetic differences in the human IL-6 gene could be linked to the risk of recurrent prostate cancer.


Asunto(s)
Interleucina-6/genética , Recurrencia Local de Neoplasia/genética , Polimorfismo Genético , Neoplasias de la Próstata/genética , Tejido Adiposo/patología , Vasos Sanguíneos/patología , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Invasividad Neoplásica , Neoplasias de la Próstata/patología , Estudios Retrospectivos , Vesículas Seminales/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA