Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Pediatr Blood Cancer ; 56(2): 191-201, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20830775

RESUMEN

BACKGROUND: (131) I-Metaiodobenzylguanidine ((131) I-MIBG) provides targeted radiotherapy for children with neuroblastoma, a malignancy of the sympathetic nervous system. Dissociated radioactive iodide may concentrate in the thyroid, and (131) I-MIBG is concentrated in the liver after (131) I-MIBG therapy. The aim of our study was to analyze the effects of (131) I-MIBG therapy on thyroid and liver function. PROCEDURE: Pre- and post-therapy thyroid and liver functions were reviewed in a total of 194 neuroblastoma patients treated with (131) I-MIBG therapy. The cumulative incidence over time was estimated for both thyroid and liver toxicities. The relationship to cumulative dose/kg, number of treatments, time from treatment to follow-up, sex, and patient age was examined. RESULTS: In patients who presented with Grade 0 or 1 thyroid toxicity at baseline, 12 ± 4% experienced onset of or worsening to Grade 2 hypothyroidism and one patient developed Grade 2 hyperthyroidism by 2 years after (131) I-MIBG therapy. At 2 years post-(131) I-MIBG therapy, 76 ± 4% patients experienced onset or worsening of hepatic toxicity to any grade, and 23 ± 5% experienced onset of or worsening to Grade 3 or 4 liver toxicity. Liver toxicity was usually transient asymptomatic transaminase elevation, frequently confounded by disease progression and other therapies. CONCLUSION: The prophylactic regimen of potassium iodide and potassium perchlorate with (131) I-MIBG therapy resulted in a low rate of significant hypothyroidism. Liver abnormalities following (131) I-MIBG therapy were primarily reversible and did not result in late toxicity. (131) I-MIBG therapy is a promising treatment for children with relapsed neuroblastoma with a relatively low rate of symptomatic thyroid or hepatic dysfunction.


Asunto(s)
3-Yodobencilguanidina/efectos adversos , Antineoplásicos/efectos adversos , Radioisótopos de Yodo/efectos adversos , Hígado/efectos de la radiación , Neuroblastoma/radioterapia , Glándula Tiroides/efectos de la radiación , Adolescente , Niño , Ensayos Clínicos como Asunto , Femenino , Humanos , Pruebas de Función Hepática , Masculino , Adulto Joven
2.
J Clin Oncol ; 27(32): 5343-9, 2009 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-19805691

RESUMEN

PURPOSE: Children with relapsed neuroblastoma have poor survival. It is crucial to have a reliable method for evaluating functional response to new therapies. In this study, we compared two functional imaging modalities for neuroblastoma: metaiodobenzylguanidine (MIBG) scan for uptake by the norepinephrine transporter and [(18)F]fluorodeoxyglucose positron emission tomography (FDG-PET) uptake for glucose metabolic activity. PATIENTS AND METHODS: Patients enrolled onto a phase I study of sequential infusion of iodine-131 ((131)I) MIBG (NANT-2000-01) were eligible for inclusion if they had concomitant FDG-PET and MIBG scans. (131)I-MIBG therapy was administered on days 0 and 14. For each patient, we compared all lesions identified on concomitant FDG-PET and MIBG scans and gave scans a semiquantitative score. RESULTS: The overall concordance of positive lesions on concomitant MIBG and FDG-PET scans was 39.6% when examining the 139 unique anatomic lesions. MIBG imaging was significantly more sensitive than FDG-PET overall and for the detection of bone lesions (P < .001). There was a trend for increased sensitivity of FDG-PET for detection of soft tissue lesions. Both modalities showed similar improvement in number of lesions identified from day 0 to day 56 scan and in semiquantitative scores that correlated with overall response. FDG-PET scans became completely negative more often than MIBG scans after treatment. CONCLUSION: MIBG scan is significantly more sensitive for individual lesion detection in relapsed neuroblastoma than FDG-PET, though FDG-PET can sometimes play a complementary role, particularly in soft tissue lesions. Complete response by FDG-PET metabolic evaluation did not always correlate with complete response by MIBG uptake.


Asunto(s)
Neuroblastoma/diagnóstico , Tomografía de Emisión de Positrones/métodos , Tomografía Computarizada por Rayos X/métodos , 3-Yodobencilguanidina , Adolescente , Niño , Preescolar , Ensayos Clínicos Fase I como Asunto , Fluorodesoxiglucosa F18 , Humanos , Radioisótopos de Yodo/uso terapéutico , Recurrencia Local de Neoplasia , Neuroblastoma/radioterapia , Evaluación de Resultado en la Atención de Salud/métodos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
3.
J Clin Oncol ; 27(25): 4162-8, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19636009

RESUMEN

PURPOSE: To evaluate the safety and efficacy of high-dose [(131)I]metaiodobenzylguanidine ([(131)I]MIBG) in the treatment of malignant pheochromocytoma (PHEO) and paraganglioma (PGL). METHODS: Fifty patients with metastatic PHEO or PGL, age 10 to 64 years, were treated with [(131)I]MIBG doses ranging from 492 to 1,160 mCi (median, 12 mCi/kg). Cumulative [(131)I]MIBG administered ranged from 492 to 3,191 mCi. Autologous hematopoietic stem cells were collected and cryopreserved before treatment with [(131)I]MIBG greater than 12 mCi/kg or with a total dose greater than 500 mCi. Sixty-nine [(131)I]MIBG infusions were given, which included infusions to 35 patients treated once and infusions to 15 patients who received two or three treatments. Response was evaluated by [(123)I]MIBG scans, computed tomography/magnetic resonance imaging, urinary catecholamines/metanephrines, and chromogranin A. RESULTS: The overall complete response (CR) plus partial response (PR) rate in 49 evaluable patients was 22%. Additionally, 35% of patients achieved a CR or PR in at least one measure of response without progressive disease, and 8% of patients maintained stable disease for greater than 12 months. Thirty-five percent of patients experienced progressive disease within 1 year after therapy. The estimated 5-year overall survival rate was 64%. Toxicities included grades 3 to 4 neutropenia (87%) and thrombocytopenia (83%). Grades 3 to 4 nonhematologic toxicity included acute respiratory distress syndrome (n = 2), bronchiolitis obliterans organizing pneumonia (n = 2), pulmonary embolism (n = 1), fever with neutropenia (n = 7), acute hypertension (n = 10), infection (n = 2), myelodysplastic syndrome (n = 2), and hypogonadism (n = 4). CONCLUSION: Although serious toxicity may occur, the survival and response rates achieved with high-dose [(131)I]MIBG suggest its utility in the management of selected patients with metastatic PHEO and PGL.


Asunto(s)
3-Yodobencilguanidina/administración & dosificación , Neoplasias de las Glándulas Suprarrenales/radioterapia , Radioisótopos de Yodo/administración & dosificación , Paraganglioma/radioterapia , Feocromocitoma/radioterapia , Radiofármacos/administración & dosificación , 3-Yodobencilguanidina/efectos adversos , Adolescente , Neoplasias de las Glándulas Suprarrenales/diagnóstico , Neoplasias de las Glándulas Suprarrenales/mortalidad , Neoplasias de las Glándulas Suprarrenales/secundario , Adulto , Biomarcadores de Tumor/orina , Catecolaminas/orina , Niño , Cromogranina A/orina , Supervivencia sin Enfermedad , Relación Dosis-Respuesta en la Radiación , Femenino , Humanos , Infusiones Intravenosas , Radioisótopos de Yodo/efectos adversos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Paraganglioma/diagnóstico , Paraganglioma/secundario , Feocromocitoma/diagnóstico , Feocromocitoma/mortalidad , Feocromocitoma/secundario , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Radiofármacos/efectos adversos , Medición de Riesgo , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
4.
J Clin Oncol ; 27(1): 85-91, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19047298

RESUMEN

PURPOSE: Recurrence of high-risk neuroblastoma is common despite multimodality therapy. ch14.18, a chimeric human/murine anti-G(D2) antibody, lyses neuroblastoma cells. This study determined the maximum tolerable dose (MTD) and toxicity of ch14.18 given in combination with interleukin-2 (IL-2) after high-dose chemotherapy (HDC)/stem-cell rescue (SCR). Biologic correlates including ch14.18 levels, soluble IL-2 receptor levels, and human antichimeric antibody (HACA) activity were evaluated. PATIENTS AND METHODS: Patients were given ch14.18 for 4 days at 28-day intervals. Patients received IL-2 during the second and fourth courses of ch14.18 and granulocyte-macrophage colony-stimulating factor (GM-CSF) during the first, third, and fifth courses. The MTD was determined based on toxicities occurring with the second course. After the determination of the MTD, additional patients were treated to confirm the MTD and to clarify appropriate supportive care. RESULTS: Twenty-five patients were enrolled. The MTD of ch14.18 was determined to be 25 mg/m(2)/d for 4 days given concurrently with 4.5 x 10(6) U/m(2)/d of IL-2 for 4 days. IL-2 was also given at a dose of 3 x 10(6) U/m(2)/d for 4 days starting 1 week before ch14.18. Two patients experienced dose-limiting toxicity due to ch14.18 and IL-2. Common toxicities included pain, fever, nausea, emesis, diarrhea, urticaria, mild elevation of hepatic transaminases, capillary leak syndrome, and hypotension. No death attributable to toxicity of therapy occurred. No additional toxicity was seen when cis-retinoic acid (cis-RA) was given between courses of ch14.18. No patient treated at the MTD developed HACA. CONCLUSION: ch14.18 in combination with IL-2 was tolerable in the early post-HDC/SCR period. cis-RA can be administered safely between courses of ch14.18 and cytokines.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Trasplante de Médula Ósea , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Trasplante de Células Madre Hematopoyéticas , Interleucina-2/uso terapéutico , Neuroblastoma/terapia , Adolescente , Anticuerpos Monoclonales/efectos adversos , Niño , Preescolar , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/efectos adversos , Humanos , Lactante , Recién Nacido , Interleucina-2/efectos adversos , Masculino
5.
Ann N Y Acad Sci ; 1073: 465-90, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17102115

RESUMEN

Thirty patients with malignant pheochromocytoma (PHEO) or paraganglioma (PGL) were treated with high-dose 131I-MIBG. Patients were 11-62 (mean 39) years old: 19 patients males and 11 females. Nineteen patients had PGL, three of which were multifocal. Six PGLs were nonsecretory. Eleven patients had PHEO. All 30 patients had prior surgery. Fourteen patients were refractory to prior radiation or chemotherapy before 131I-MIBG. Peripheral blood stem cells (PBSCs) were collected and cryopreserved. 131I-MIBG was synthesized on-site, by exchange-labeling 131I with 127I-MIBG in a solid-phase Cu2+-catalyzed exchange reaction. 131I-MIBG was infused over 2 h via a peripheral IV. Doses ranged from 557 mCi to 1185 mCi (7.4 mCi/kg to 18.75 mCi/kg). Median dose was 833 mCi (12.55 mCi/kg). Marrow hypoplasia commenced 3 weeks after 131I-MIBG therapy. After the first 131I-MIBG therapy, 19 patients required platelet transfusions; 19 received GCSF; 12 received epoeitin or RBCs. Four patients received a PBSC infusion. High-dose 131I-MIBG resulted in the following overall tumor responses in 30 patients: 4 sustained complete remissions (CRs); 15 sustained partial remissions (PRs); 1 sustained stable disease (SD); 5 progressive disease (PD); 5 initial PRs or SD but relapsed to PD. Twenty-three of the 30 patients remain alive; deaths were from PD (5), myelodysplasia (1), and unrelated cause (1). Overall predicted survival at 5 years is 75% (Kaplan Meier estimate). For patients with metastatic PHEO or PGL, who have good *I-MIBG uptake on diagnostic scanning, high-dose 131I-MIBG therapy was effective in producing a sustained CR, PR, or SD in 67% of patients, with tolerable toxicity.


Asunto(s)
3-Yodobencilguanidina/uso terapéutico , Neoplasias de las Glándulas Suprarrenales/radioterapia , Radioisótopos de Yodo/uso terapéutico , Paraganglioma/radioterapia , Feocromocitoma/radioterapia , 3-Yodobencilguanidina/efectos adversos , Adolescente , Neoplasias de las Glándulas Suprarrenales/diagnóstico , Neoplasias de las Glándulas Suprarrenales/metabolismo , Adulto , Niño , Diagnóstico Diferencial , Relación Dosis-Respuesta en la Radiación , Femenino , Humanos , Radioisótopos de Yodo/efectos adversos , Masculino , Persona de Mediana Edad , Paraganglioma/diagnóstico , Feocromocitoma/diagnóstico , Feocromocitoma/metabolismo
6.
J Clin Oncol ; 23(27): 6474-80, 2005 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-16116154

RESUMEN

PURPOSE: The long-term survival of children between age 12 and 24 months with stage 4 neuroblastoma and nonamplified MYCN (MYCN-NA) has not been defined previously. PATIENTS AND METHODS: Survival for stage 4 MYCN-NA neuroblastoma patients enrolled onto Children's Cancer Group (CCG) protocols 321P2 (1986 to 1991) and 3891 (1991 to 1996) was analyzed. Treatment consisted of intensive alkylator-based induction chemotherapy with or without autologous bone marrow transplantation (ABMT) with or without 13 cis-retinoic acid. Survival was analyzed by age strata less than 12, 12 to 18, 18 to 24, and more than 24 months at diagnosis. Patients younger than 12 months were treated on the moderate-intensity CCG protocol 3881. RESULTS: Forty-three patients with stage 4 MYCN-NA disease enrolled onto CCG-321P2 (n = 17) or CCG-3891 (n = 26) were between 12 and 24 months of age at diagnosis. After a median follow-up of 94 months (range, 4 to 140 months), the 6-year event-free survival (EFS) for the 12- to 18-month age group was superior to that of the 18- to 24-month age group (74% +/- 8% v 31% +/- 12%; P = .008). The EFS for children older than 24 months with stage 4 MYCN-NA neuroblastoma was 23% +/- 3%, and for children younger than 12 months was 92% +/- 3%. CONCLUSION: Children diagnosed with stage 4 MYCN-NA neuroblastoma in the second year of life form a transitional group between infants and older children in terms of prognosis. Patients between 12 and 18 months of age have significantly better long-term survival than that of older children treated with intensive chemotherapy with or without ABMT. These patients may not benefit from additional intensification of therapy beyond that provided in earlier clinical trials and may even maintain this high survival rate with less intensive therapy.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Genes myc/genética , Neuroblastoma/mortalidad , Neuroblastoma/patología , Factores de Edad , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Trasplante de Médula Ósea , Preescolar , Terapia Combinada , Supervivencia sin Enfermedad , Femenino , Amplificación de Genes , Humanos , Lactante , Masculino , Estadificación de Neoplasias , Neuroblastoma/genética , Neuroblastoma/terapia , Pronóstico , Radioterapia Adyuvante , Medición de Riesgo , Procedimientos Quirúrgicos Operativos/métodos , Análisis de Supervivencia , Resultado del Tratamiento
7.
Pediatr Blood Cancer ; 44(3): 232-9, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15490487

RESUMEN

BACKGROUND: (131)I Metaiodobenzylguanidine ((131)I-MIBG) is an effective targeted radiotherapeutic for neuroblastoma with response rates greater than 30% in refractory disease. Toxicity is mainly limited to myelosuppression. The aim of this study was to determine the response rate and hematologic toxicity of multiple infusions of (131)I-MIBG. PROCEDURE: Patients received two to four infusions of (131)I-MIBG at activity levels of 3-19 mCi/kg per infusion. Criteria for subsequent infusions were neutrophil recovery without stem cell support and lack of disease progression after the first infusion. RESULTS: Sixty-two infusions were administered to 28 patients, with 24 patients receiving two infusions, two patients receiving three infusions, and two patients receiving four infusions. All patients were heavily pre-treated, including 16 with prior myeloablative therapy. Eleven patients (39%) had overall disease response to multiple therapies, including eight patients with measurable responses to each of two or three infusions, and three with a partial response (PR) after the first infusion and stable disease after the second. The main toxicity was myelosuppression, with 78% and 82% of patients requiring platelet transfusion support after the first and second infusion, respectively, while only 50% had grade 4 neutropenia, usually transient. Thirteen patients did not recover platelet transfusion independence after their final MIBG infusion; stem cell support was given in ten patients. CONCLUSIONS: Multiple therapies with (131)I-MIBG achieved increasing responses, but hematologic toxicity, especially to platelets, was dose limiting. More effective therapy might be given using consecutive doses in rapid succession with early stem cell support.


Asunto(s)
3-Yodobencilguanidina/administración & dosificación , 3-Yodobencilguanidina/toxicidad , Médula Ósea/efectos de los fármacos , Radioisótopos de Yodo/administración & dosificación , Radioisótopos de Yodo/toxicidad , Neuroblastoma/radioterapia , Radiofármacos/administración & dosificación , Radiofármacos/toxicidad , Adolescente , Adulto , Anemia/inducido químicamente , Niño , Preescolar , Supervivencia sin Enfermedad , Femenino , Humanos , Infusiones Intravenosas , Masculino , Neutropenia/inducido químicamente , Trombocitopenia/inducido químicamente , Resultado del Tratamiento
8.
J Clin Oncol ; 22(12): 2452-60, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15197208

RESUMEN

PURPOSE: Iodine-131-metaiodobenzylguanidine ((131)I-MIBG) has been shown to be active against refractory neuroblastoma. The primary toxicity of (131)I-MIBG is myelosuppression, which might necessitate autologous hematopoietic stem-cell transplantation (AHSCT). The goal of this study was to determine risk factors for myelosuppression and the need for AHSCT after (131)I-MIBG treatment. PATIENTS AND METHODS: Fifty-three patients with refractory or relapsed neuroblastoma were treated with 18 mCi/kg (131)I-MIBG on a phase I/II protocol. The median whole-body radiation dose was 2.92 Gy. RESULTS: Almost all patients required at least one platelet (96%) or red cell (91%) transfusion and most patients (79%) developed neutropenia (< 0.5 x 10(3)/microL). Patients reached platelet nadir earlier than neutrophil nadir (P <.0001). Earlier platelet nadir correlated with bone marrow tumor, more extensive bone involvement, higher whole-body radiation dose, and longer time from diagnosis to (131)I-MIBG therapy (P

Asunto(s)
3-Yodobencilguanidina/administración & dosificación , 3-Yodobencilguanidina/efectos adversos , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Neuroblastoma/tratamiento farmacológico , Radiofármacos/administración & dosificación , Radiofármacos/efectos adversos , Adolescente , Adulto , Niño , Preescolar , Femenino , Trasplante de Células Madre Hematopoyéticas , Humanos , Lactante , Radioisótopos de Yodo/uso terapéutico , Masculino , Neutropenia/inducido químicamente , Neutropenia/terapia , Trombocitopenia/inducido químicamente , Trombocitopenia/terapia , Trasplante Autólogo
9.
Cancer Lett ; 197(1-2): 185-92, 2003 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-12880980

RESUMEN

Retinoids are derivatives of vitamin A that include all trans-retinoic acid (ATRA), 13-cis-retinoic acid, (13-cis-RA), and fenretinide (4-HPR). High levels of either ATRA or 13-cis-RA can cause arrest of cell growth and morphological differentiation of human neuroblastoma cell lines, and phase I trials showed that higher and more sustained drug levels were obtained with 13-cis-RA relative to ATRA. A phase III randomized trial showed that high-dose, pulse therapy with 13-cis-RA given after completion of intensive chemoradiotherapy (with or without autologous bone marrow transplantation) significantly improved event-free survival in high-risk neuroblastoma. The cytotoxic retinoid 4-HPR achieved multi-log cell kills in neuroblastoma cell lines resistant to ATRA and 13-cis-RA, and a pediatric phase I trial has shown it to be well tolerated. Cytotoxicity of 4-HPR is mediated at least in part by increasing tumor cell ceramide levels and combining 4-HPR with ceramide modulators increased anti-tumor activity in pre-clinical models. Thus, further clinical trials of 4-HPR in neuroblastoma, and of 4-HPR in combination with ceramide modulators, are warranted.


Asunto(s)
Antineoplásicos/uso terapéutico , Neuroblastoma/tratamiento farmacológico , Tretinoina/uso terapéutico , Ensayos Clínicos como Asunto , Humanos , Receptores de Ácido Retinoico/efectos de los fármacos , Receptores de Ácido Retinoico/metabolismo
10.
Int J Radiat Oncol Biol Phys ; 56(1): 28-39, 2003 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-12694821

RESUMEN

PURPOSE: To assess the effect of local radiation administered to primary disease sites in children with high-risk neuroblastoma. MATERIALS AND METHODS: A total of 539 eligible patients were entered on protocol CCG-3891, consisting of chemotherapy, primary surgery, and 10 Gy of external beam radiation therapy (EBRT) to gross residual disease, followed by randomized assignment to continuation chemotherapy (CC) or autologous bone marrow transplantation (ABMT). ABMT patients received total body irradiation (TBI). RESULTS: Estimated event-free survival and overall survival at 5 years were 25% +/- 2% and 35% +/- 2%, respectively. Estimated 5-year locoregional recurrence rates were 51% +/- 5% and 33% +/- 7% for CC and ABMT patients (p = 0.004). For patients who received 10 Gy of EBRT to the primary, the addition of 10 Gy of TBI and ABMT decreased local recurrence compared with CC (22% +/- 12% and 52% +/- 8%, p = 0.022). EBRT did not increase acute toxicity, except for increased total parenteral nutrition administration. CONCLUSIONS: In combination with EBRT to the primary tumor site, the addition of 10 Gy of TBI as a component of high-dose chemotherapy with ABMT improved local control compared with CC without TBI. Results suggest a dose-response relationship for local EBRT. Short-term toxicity of local EBRT is limited.


Asunto(s)
Neuroblastoma/radioterapia , Radioterapia Adyuvante , Irradiación Corporal Total , Adolescente , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Purgación de la Médula Ósea , Trasplante de Médula Ósea , Niño , Preescolar , Cisplatino/administración & dosificación , Terapia Combinada , Ciclofosfamida/administración & dosificación , Fraccionamiento de la Dosis de Radiación , Doxorrubicina/administración & dosificación , Enfermedades Hematológicas/inducido químicamente , Enfermedad Veno-Oclusiva Hepática/etiología , Humanos , Lactante , Isotretinoína/uso terapéutico , Tablas de Vida , Recurrencia Local de Neoplasia , Neoplasias Inducidas por Radiación/etiología , Neoplasias Primarias Secundarias/epidemiología , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/cirugía , Neuroblastoma/terapia , Radioterapia Adyuvante/efectos adversos , Inducción de Remisión , Riesgo , Análisis de Supervivencia , Tasa de Supervivencia , Trasplante Autólogo , Irradiación Corporal Total/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA