Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Pediatr Blood Cancer ; 63(8): 1349-56, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27092812

RESUMEN

BACKGROUND: Myeloablative therapy for high-risk neuroblastoma commonly includes melphalan. Increased cellular glutathione (GSH) can mediate melphalan resistance. Buthionine sulfoximine (BSO), a GSH synthesis inhibitor, enhances melphalan activity against neuroblastoma cell lines, providing the rationale for a Phase 1 trial of BSO-melphalan. PROCEDURES: Patients with recurrent/resistant high-risk neuroblastoma received BSO (3 gram/m(2) bolus, then 24 grams/m(2) /day infusion days -4 to -2), with escalating doses of intravenous melphalan (20-125 mg/m(2) ) days -3 and -2, and autologous stem cells day 0 using 3 + 3 dose escalation. RESULTS: Among 28 patients evaluable for dose escalation, one dose-limiting toxicity occurred at 20 mg/m(2) melphalan (grade 3 aspartate aminotransferase/alanine aminotransferase) and one at 80 mg/m(2) (streptococcal bacteremia, grade 4 hypotension/pulmonary/hypocalcemia) without sequelae. Among 25 patients evaluable for response, there was one partial response (PR) and two mixed responses (MRs) among eight patients with prior melphalan exposure; one PR and three MRs among 16 patients without prior melphalan; one stable disease with unknown melphalan history. Melphalan pharmacokinetics with BSO were similar to reports for melphalan alone. Melphalan Cmax for most patients was below the 10 µM concentration that showed neuroblastoma preclinical activity with BSO. CONCLUSIONS: BSO (75 gram/m(2) ) with melphalan (125 mg/m(2) ) is tolerable with stem cell support and active in recurrent/refractory neuroblastoma. Further dose escalation is feasible and may increase responses.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Antineoplásicos Alquilantes/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Butionina Sulfoximina/uso terapéutico , Trasplante de Células Madre Hematopoyéticas , Melfalán/uso terapéutico , Agonistas Mieloablativos/uso terapéutico , Neuroblastoma/tratamiento farmacológico , Adolescente , Butionina Sulfoximina/efectos adversos , Niño , Preescolar , Sinergismo Farmacológico , Femenino , Glutamato-Cisteína Ligasa/antagonistas & inhibidores , Glutatión/uso terapéutico , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Melfalán/efectos adversos , Melfalán/farmacocinética , Recurrencia Local de Neoplasia/tratamiento farmacológico
2.
Biol Blood Marrow Transplant ; 21(4): 673-81, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25639769

RESUMEN

(131)I-Metaiodobenzylguanidine ((131)I-MIBG) has been used as a single agent or in combination with chemotherapy for the treatment of high-risk neuroblastoma. The activity and toxicity of (131)I-MIBG when combined with carboplatin, etoposide, and melphalan (CEM) and autologous stem cell transplantation (SCT) are now investigated in a phase II multicenter study. Fifty patients with MIBG-avid disease were enrolled into 2 cohorts, stratified by response to induction therapy. The primary study endpoint was response of patients with refractory (n = 27) or progressive disease (n = 15). A second cohort of patients (n = 8) with a partial response (PR) to induction therapy was included to obtain preliminary response data. (131)I-MIBG was administered on day -21 to all patients, with CEM given days -7 to -4, and SCT given on day 0. (131)I-MIBG dosing was determined by pre-therapy glomerular filtration rate (GFR), with 8 mCi/kg given if GFR was 60 to 99 mL/minute/1.73 m(2) (n = 13) and 12 mCi/kg if GFR ≥ 100 mL/minute/1.73 m(2) (n = 37). External beam radiotherapy was delivered to the primary and metastatic sites, beginning approximately 6 weeks after SCT. Responses (complete response + PR) were seen in 4 of 41 (10%) evaluable patients with primary refractory or progressive disease. At 3 years after SCT, the event-free survival (EFS) was 20% ± 7%, with overall survival (OS) 62% ± 8% for this cohort of patients. Responses were noted in 3 of 8 (38%) of patients with a PR to induction, with 3-year EFS 38% ± 17% and OS 75% ± 15%. No statistically significant difference was found comparing EFS or OS based upon pre-therapy GFR or disease cohort. Six of 50 patients had nonhematologic dose-limiting toxicity (DLT); 1 of 13 in the low GFR and 5 of 37 in the normal GFR cohorts. Hepatic sinusoidal obstructive syndrome (SOS) was seen in 6 patients (12%), with 5 events defined as dose-limiting SOS. The median times to neutrophil and platelet engraftment were 10 and 15 days, respectively. Patients received a median 163 cGy (61 to 846 cGy) with (131)I-MIBG administration, with 2 of 3 patients receiving >500 cGy experiencing DLT. The addition of (131)I-MIBG to a myeloablative CEM regimen is tolerable and active therapy for patients with high-risk neuroblastoma.


Asunto(s)
3-Yodobencilguanidina/administración & dosificación , Antineoplásicos/administración & dosificación , Neuroblastoma/terapia , Trasplante de Células Madre , Adulto , Autoinjertos , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Neuroblastoma/mortalidad , Terapia de Protones
3.
N Engl J Med ; 363(14): 1324-34, 2010 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-20879881

RESUMEN

BACKGROUND: Preclinical and preliminary clinical data indicate that ch14.18, a monoclonal antibody against the tumor-associated disialoganglioside GD2, has activity against neuroblastoma and that such activity is enhanced when ch14.18 is combined with granulocyte-macrophage colony-stimulating factor (GM-CSF) or interleukin-2. We conducted a study to determine whether adding ch14.18, GM-CSF, and interleukin-2 to standard isotretinoin therapy after intensive multimodal therapy would improve outcomes in high-risk neuroblastoma. METHODS: Patients with high-risk neuroblastoma who had a response to induction therapy and stem-cell transplantation were randomly assigned, in a 1:1 ratio, to receive standard therapy (six cycles of isotretinoin) or immunotherapy (six cycles of isotretinoin and five concomitant cycles of ch14.18 in combination with alternating GM-CSF and interleukin-2). Event-free survival and overall survival were compared between the immunotherapy group and the standard-therapy group, on an intention-to-treat basis. RESULTS: A total of 226 eligible patients were randomly assigned to a treatment group. In the immunotherapy group, a total of 52% of patients had pain of grade 3, 4, or 5, and 23% and 25% of patients had capillary leak syndrome and hypersensitivity reactions, respectively. With 61% of the number of expected events observed, the study met the criteria for early stopping owing to efficacy. The median duration of follow-up was 2.1 years. Immunotherapy was superior to standard therapy with regard to rates of event-free survival (66±5% vs. 46±5% at 2 years, P=0.01) and overall survival (86±4% vs. 75±5% at 2 years, P=0.02 without adjustment for interim analyses). CONCLUSIONS: Immunotherapy with ch14.18, GM-CSF, and interleukin-2 was associated with a significantly improved outcome as compared with standard therapy in patients with high-risk neuroblastoma. (Funded by the National Institutes of Health and the Food and Drug Administration; ClinicalTrials.gov number, NCT00026312.)


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Gangliósidos/inmunología , Inmunoterapia , Neuroblastoma/terapia , Anticuerpos Monoclonales/efectos adversos , Antineoplásicos/uso terapéutico , Niño , Preescolar , Terapia Combinada , Quimioterapia Combinada , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Humanos , Inmunoterapia/efectos adversos , Lactante , Análisis de Intención de Tratar , Interleucina-2/uso terapéutico , Isotretinoína/uso terapéutico , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/mortalidad , Trasplante de Células Madre , Análisis de Supervivencia
4.
J Microbiol Biotechnol ; 26(5): 918-27, 2016 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-26437944

RESUMEN

Cryptococcus neoformans is a life-threatening pathogenic yeast that causes devastating meningoencephalitis. The mechanism of cryptococcal brain invasion is largely unknown, and recent studies suggest that its extracellular microvesicles may be involved in the invasion process. The 14-3-3 protein is abundant in the extracellular microvesicles of C. neoformans, and the 14-3-3-GFP fusion has been used as the microvesicle's marker. However, the physiological role of 14-3-3 has not been explored. In this report, we have found that C. neoformans contains a single 14-3-3 gene that apparently is an essential gene. To explore the functions of 14-3-3, we substituted the promoter region of the 14-3-3 with the copper-controllable promoter CTR4. The CTR4 regulatory strain showed an enlarged cell size, drastic changes in morphology, and a decrease in the thickness of the capsule under copper-enriched conditions. Furthermore, the mutant cells produced a lower amount of total proteins in their extracellular microvesicles and reduced adhesion to human brain microvascular endothelial cells in vitro. Proteomic analyses of the protein components under 14-3-3-overexpressed and -suppressed conditions revealed that the 14-3-3 function(s) might be associated with the microvesicle biogenesis. Our results support that 14-3-3 has diverse pertinent roles in both physiology and pathogenesis in C. neoformans. Its gene functions are closely relevant to the pathogenesis of this fungus.


Asunto(s)
Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Cryptococcus neoformans/genética , Fosfatasa Ácida/metabolismo , Biomarcadores/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/microbiología , Adhesión Celular/fisiología , Cobre/metabolismo , Cryptococcus neoformans/crecimiento & desarrollo , Cryptococcus neoformans/patogenicidad , Células Endoteliales/microbiología , Cápsulas Fúngicas/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Humanos , Lacasa/metabolismo , Mutación , Fenotipo , Regiones Promotoras Genéticas , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
5.
Cancer Res ; 68(16): 6507-15, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18701473

RESUMEN

Deregulation of the polycomb group gene BMI-1 is implicated in the pathogenesis of many human cancers. In this study, we have investigated if the Ewing sarcoma family of tumors (ESFT) expresses BMI-1 and whether it functions as an oncogene in this highly aggressive group of bone and soft tissue tumors. Our data show that BMI-1 is highly expressed by ESFT cells and that, although it does not significantly affect proliferation or survival, BMI-1 actively promotes anchorage-independent growth in vitro and tumorigenicity in vivo. Moreover, we find that BMI-1 promotes the tumorigenicity of both p16 wild-type and p16-null cell lines, demonstrating that the mechanism of BMI-1 oncogenic function in ESFT is, at least in part, independent of CDKN2A repression. Expression profiling studies of ESFT cells following BMI-1 knockdown reveal that BMI-1 regulates the expression of hundreds of downstream target genes including, in particular, genes involved in both differentiation and development as well as cell-cell and cell-matrix adhesion. Gain and loss of function assays confirm that BMI-1 represses the expression of the adhesion-associated basement membrane protein nidogen 1. In addition, although BMI-1 promotes ESFT adhesion, nidogen 1 inhibits cellular adhesion in vitro. Together, these data support a pivotal role for BMI-1 ESFT pathogenesis and suggest that its oncogenic function in these tumors is in part mediated through modulation of adhesion pathways.


Asunto(s)
Neoplasias Óseas/patología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Proteínas Nucleares/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Represoras/fisiología , Sarcoma de Ewing/patología , Animales , Western Blotting , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Adhesión Celular , Proliferación Celular , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Nucleares/antagonistas & inhibidores , Análisis de Secuencia por Matrices de Oligonucleótidos , Complejo Represivo Polycomb 1 , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , ARN Interferente Pequeño/farmacología , Proteínas Represoras/antagonistas & inhibidores , Sarcoma de Ewing/genética , Sarcoma de Ewing/metabolismo , Análisis de Matrices Tisulares
6.
Conf Proc IEEE Eng Med Biol Soc ; 2006: 4844-7, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17947119

RESUMEN

Neuroblastoma is a childhood cancer of the nervous system. Current prognostic classification of this disease partly relies on morphological characteristics of the cells from H&E-stained images. In this work, an automated cell nuclei segmentation method is developed. This method employs morphological top-hat by reconstruction algorithm coupled with hysteresis thresholding to both detect and segment the cell nuclei. Accuracy of the automated cell nuclei segmentation algorithm is measured by comparing its outputs to manual segmentation. The average segmentation accuracy is 90.24+/-5.14%


Asunto(s)
Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/patología , Núcleo Celular/metabolismo , Procesamiento de Imagen Asistido por Computador/instrumentación , Procesamiento de Imagen Asistido por Computador/métodos , Oncología Médica/instrumentación , Neuroblastoma/diagnóstico , Neuroblastoma/patología , Algoritmos , Automatización , Diagnóstico Diferencial , Diseño de Equipo , Humanos , Interpretación de Imagen Asistida por Computador/instrumentación , Oncología Médica/métodos , Reconocimiento de Normas Patrones Automatizadas , Pronóstico , Reproducibilidad de los Resultados
7.
Transl Res ; 148(5): 272-80, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17145573

RESUMEN

INTRODUCTION: Deferasirox effectively controls liver iron concentration; however, little is known regarding its ability to remove stored cardiac iron. Deferiprone seems to have increased cardiac efficacy compared with traditional deferoxamine therapy. Therefore, the relative efficacy of deferasirox and deferiprone were compared in removing cardiac iron from iron-loaded gerbils. METHODS: Twenty-nine 8- to 10-week-old female gerbils underwent 10 weekly iron dextran injections of 200 mg/kg/week. Prechelation iron levels were assessed in 5 animals, and the remainder received deferasirox 100 mg/kg/D po QD (n = 8), deferiprone 375 mg/kg/D po divided TID (n = 8), or sham chelation (n = 8), 5 days/week for 12 weeks. RESULTS: Deferasirox reduced cardiac iron content 20.5%. No changes occurred in cardiac weight, myocyte hypertrophy, fibrosis, or weight-to-dry weight ratio. Deferasirox treatment reduced liver iron content 51%. Deferiprone produced comparable reductions in cardiac iron content (18.6% reduction). Deferiprone-treated hearts had greater mass (16.5% increase) and increased myocyte hypertrophy. Deferiprone decreased liver iron content 24.9% but was associated with an increase in liver weight and water content. CONCLUSION: Deferasirox and deferiprone were equally effective in removing stored cardiac iron in a gerbil animal model, but deferasirox removed more hepatic iron for a given cardiac iron burden.


Asunto(s)
Benzoatos/uso terapéutico , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Piridonas/uso terapéutico , Triazoles/uso terapéutico , Animales , Deferasirox , Deferiprona , Modelos Animales de Enfermedad , Femenino , Gerbillinae , Corazón/efectos de los fármacos , Hierro/metabolismo , Sobrecarga de Hierro/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Tamaño de los Órganos/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA