Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Pediatr Hematol Oncol ; 37(3): e184-7, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25171448

RESUMEN

Peutz-Jeghers syndrome (PJS) is a rare autosomal dominant disorder that typically displays familial inheritance. Gastrointestinal polyposis and cutaneous pigmentation is a classic presentation of this syndrome. The reported lifetime cumulative cancer risk in PJS patients is >76% when compared with the general public with females affected more often than males. The prepubertal testicular tumor registry found Sertoli cell tumors (SCTs) to compose approximately 1% of all pediatric solid tumors. Prepubertal testicular masses are relatively rare. Only a small number of SCT cases have been reported in the first decade of life. The concurrence of PJS and feminizing SCTs of the testes is an increasingly recognized cause of prepubertal gynecomastia. The testicular lesions observed in patients with PJS primarily represent multifocal intratubular large cell hyalinizing SCTs with a distinct morphology that differs from large cell calcifying SCTs and sex cord tumors with annular tubules. Here, we describe the diagnosis and treatment course of a 4-year-old male with a SCT of the testes and diagnosis of PJS.


Asunto(s)
Ginecomastia/patología , Síndrome de Peutz-Jeghers/patología , Tumor de Células de Sertoli/patología , Neoplasias Testiculares/patología , Preescolar , Femenino , Ginecomastia/etiología , Ginecomastia/cirugía , Humanos , Masculino , Síndrome de Peutz-Jeghers/complicaciones , Síndrome de Peutz-Jeghers/cirugía , Pronóstico , Tumor de Células de Sertoli/etiología , Tumor de Células de Sertoli/cirugía , Neoplasias Testiculares/etiología , Neoplasias Testiculares/cirugía
2.
J Neurooncol ; 109(3): 565-71, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22790443

RESUMEN

A pilot study to investigate the feasibility of the addition of intrathecal (IT) mafosfamide to a regimen of concomitant multi-agent systemic chemotherapy followed by conformal radiation therapy (RT) for children <3 years with newly diagnosed embryonal CNS tumors was performed. Ninety-three newly diagnosed infants and children (<3 years) with embryonal CNS tumors were enrolled. Twenty weeks of systemic multi-agent chemotherapy commenced within 35 days of surgery. Patients without CSF flow obstruction (n = 71) received IT mafosfamide (14 mg) with chemotherapy. Localized (M(0)) patients with SD or better subsequently received RT followed by 20 additional weeks of chemotherapy. Second look surgery was encouraged prior to RT if there was an incomplete surgical resection at diagnosis. 71 evaluable patients with normal CSF flow received IT Mafosfamide with systemic chemotherapy; patients with M + disease were removed from protocol therapy at 20 weeks and those with PD at the time of progression. One and 5-year progression free survival (PFS) and overall survival (OS) for the cohort of 71 evaluable patients were 52 ± 6.5 % and 33 ± 13 %, and 67 ± 6.2 % and 51 ± 11 %, respectively. The 1-year Progression Free Survival (PFS) for M0 patients with medulloblastoma (MB, n = 20), supratentorial primitive neuroectodermal tumor (PNET, n = 9), and atypical teratoid rhabdoid tumor (ATRT, n = 12) was 80 ± 7 %, 67 ± 15 % and 27 ± 13 % and 5-year PFS was 65 ± 19 %, 37 ± 29 %, and 0 ± 0 %, respectively. The addition of IT mafosfamide to systemic chemotherapy in infants with embryonal CNS tumors was feasible. The PFS for M0 patients appears comparable to or better than most prior historical comparisons and was excellent for those receiving conformal radiotherapy.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia , Ciclofosfamida/análogos & derivados , Antineoplásicos/efectos adversos , Neoplasias Encefálicas/patología , Ciclofosfamida/administración & dosificación , Ciclofosfamida/efectos adversos , Supervivencia sin Enfermedad , Estudios de Factibilidad , Femenino , Humanos , Lactante , Inyecciones Espinales , Masculino , Estadificación de Neoplasias , Proyectos Piloto , Radioterapia Conformacional
3.
Pediatr Blood Cancer ; 54(7): 909-15, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20405511

RESUMEN

BACKGROUND: The combination of irinotecan, temozolomide, and vincristine is appealing because of potentially synergistic mechanisms of action and non-overlapping toxicities. This phase I study was designed to determine the toxicity and maximum tolerated dose (MTD) of escalating daily protracted doses of irinotecan given in this combination. With extended accrual, we more fully explored the toxicity of multiple courses at the MTD. PROCEDURE: Patients under 22 years with recurrent or refractory solid tumors were eligible. A course of chemotherapy was given every 28 days. Cefpodoxime was given for diarrhea prophylaxis. Vincristine (1.5 mg/m2, max 2 mg) was given intravenously (IV) on days 1 and 8. Temozolomide (100 mg/m2/day) was given orally on days 1-5. Irinotecan was given IV over 1 hr on days 1-5 and 8-12. Dose escalation was done in the standard 3 + 3 cohort design, starting at 15 mg/m2/day. RESULTS: Twenty-five of 26 eligible patients were evaluable for toxicity and response. They received 111 courses (1-13, median 4). Dose limiting toxicity (DLT-pancreatitis, transaminitis) was seen in two of three patients at dose level 2 (20 mg/m2). No patients at level 1 had DLT during the first two cycles. Thus, the MTD of irinotecan in this combination is 15 mg/m2/day x 10 doses. Hematologic toxicity was mild and not prolonged. Grade 3 diarrhea was seen in five courses. Responses included two complete and two partial with 12 stable disease (SD) (median 6 months). CONCLUSIONS: This combination is safe and shows activity in pediatric patients with recurrent malignancy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias/tratamiento farmacológico , Adolescente , Antibióticos Antineoplásicos , Camptotecina/administración & dosificación , Camptotecina/efectos adversos , Camptotecina/análogos & derivados , Niño , Preescolar , Dacarbazina/administración & dosificación , Dacarbazina/efectos adversos , Dacarbazina/análogos & derivados , Femenino , Humanos , Lactante , Irinotecán , Masculino , Dosis Máxima Tolerada , Temozolomida , Vincristina/administración & dosificación , Vincristina/efectos adversos , Adulto Joven
4.
Am J Med Genet A ; 146A(20): 2589-97, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18798318

RESUMEN

Curry-Jones syndrome (OMIM #601707) is a rare multiple malformation disorder of unknown etiology, associated with brain and skull abnormalities, polysyndactyly, and defects of the eyes, skin and gastrointestinal tract. We report on two new cases of Curry-Jones syndrome with previously unreported features, including benign and malignant neoplasms. The first patient had typical features of Curry-Jones syndrome as well as multiple intra-abdominal smooth muscle hamartomas and trichoblastoma of the skin. The second patient was born with occipital meningoceles and developed a desmoplastic medulloblastoma. Routine lymphocyte karyotype, GLI3 gene analysis and Patched (PTCH) gene analysis on both patients and chromosome microarray analysis on the first patient were normal. We review the previously reported cases of Curry-Jones syndrome and compare our patients' findings. In view of the association of trichoblastoma with basal cell carcinoma and desmoplastic medulloblastoma with nevoid basal cell carcinoma syndrome (NBCCS) and PTCH mutations, we hypothesize that Curry-Jones syndrome is caused by malfunction of an element in the sonic hedgehog pathway.


Asunto(s)
Anomalías Múltiples , Meduloblastoma , Neoplasias Cutáneas , Anomalías Múltiples/etiología , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Agenesia del Cuerpo Calloso , Neoplasias Encefálicas/etiología , Neoplasias Encefálicas/patología , Preescolar , Coloboma/etiología , Coloboma/patología , Femenino , Tracto Gastrointestinal/anomalías , Hamartoma/etiología , Hamartoma/patología , Humanos , Hidrocefalia/etiología , Lactante , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Meduloblastoma/etiología , Meduloblastoma/patología , Meningocele/etiología , Meningocele/patología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptores Patched , Receptor Patched-1 , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/patología , Cráneo/anomalías , Sindactilia/etiología , Sindactilia/patología , Síndrome , Proteína Gli3 con Dedos de Zinc
5.
Pediatr Blood Cancer ; 51(1): 53-8, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18266227

RESUMEN

BACKGROUND: Catheter-associated blood stream infections (CABSI) are frequent complications encountered with cancer treatment. In order to understand which factors might predispose to CABSIs in children and young adults, we evaluated risk for infection in association with tumor type, catheter type, and setting of occurrence. METHODS: All pediatric oncology patients having a central venous catheter (CVC) with a tunneled external (TE) or totally implantable design (TID) were prospectively followed for the occurrence of a CABSI for 12 months. CABSIs were defined in accordance with the guidelines published by the Centers for Disease Control, and were quantified as the number of occurrences per 1,000 device days. Rates of CABSIs were stratified by tumor histology, type of catheter design, and setting of occurrence. Statistical comparisons were made using the Mantel-Haenzel statistic and the Cox proportional hazard model. RESULTS: A total of 58 CABSIs were identified in 139 patients over a period of 35,935 CVC days. The overall CABSI rate was 1.6 infections per 1,000 CVC days (95% CI 1.2, 2.1). Stratified analysis demonstrated increased risk for CABSIs in hospitalized patients having TEs, and while patients with solid tumors were also at higher risk; this association was not supported by the Cox proportional hazard model. CONCLUSION: While our baseline CABSI rate was comparatively lower than for other institutions, subset analyses identified that hospitalized cancer patients having TEs are at the highest risk for developing CABSIs. Our findings may help to guide improved methods of anticipating and controlling infections in immunocompromised patients.


Asunto(s)
Bacteriemia/etiología , Cateterismo Venoso Central/efectos adversos , Catéteres de Permanencia/efectos adversos , Neoplasias/complicaciones , Adolescente , Adulto , Cateterismo Venoso Central/instrumentación , Niño , Preescolar , Diseño de Equipo , Femenino , Humanos , Incidencia , Lactante , Masculino , Factores de Riesgo , Sepsis
6.
J Clin Oncol ; 23(7): 1555-63, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15735131

RESUMEN

PURPOSE: Preclinical studies of mafosfamide, a preactivated cyclophosphamide analog, were performed to define a tolerable and potentially active target concentration for intrathecal (IT) administration. A phase I and pharmacokinetic study of IT mafosfamide was performed to determine a dose for subsequent phase II trials. PATIENTS AND METHODS: In vitro cytotoxicity studies were performed in MCF-7, Molt-4, and rhabdomyosarcoma cell lines. Feasibility and pharmacokinetic studies were performed in nonhuman primates. These preclinical studies were followed by a phase I trial in patients with neoplastic meningitis. There were five dose levels ranging from 1 mg to 6.5 mg. Serial CSF samples were obtained for pharmacokinetic studies in a subset of patients with Ommaya reservoirs. RESULTS: The cytotoxic target exposure for mafosfamide was 10 micromol/L. Preclinical studies demonstrated that this concentration could be easily achieved in ventricular CSF after intraventricular dosing. In the phase I clinical trial, headache was the dose-limiting toxicity. Headache was ameliorated at 5 mg by prolonging the infusion rate to 20 minutes, but dose-limiting headache occurred at 6.5 mg dose with prolonged infusion. Ventricular CSF mafosfamide concentrations at 5 mg exceeded target cytotoxic concentrations after an intraventricular dose, but lumbar CSF concentrations 2 hours after the dose were less than 10 micromol/L. Therefore, a strategy to alternate dosing between the intralumbar and intraventricular routes was tested. Seven of 30 registrants who were assessable for response had a partial response, and six had stable disease. CONCLUSION: The recommended phase II dose for IT mafosfamide, administered without concomitant analgesia, is 5 mg over 20 minutes.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Ciclofosfamida/análogos & derivados , Ciclofosfamida/administración & dosificación , Ciclofosfamida/farmacocinética , Animales , Antineoplásicos/líquido cefalorraquídeo , Antineoplásicos/toxicidad , Preescolar , Ciclofosfamida/líquido cefalorraquídeo , Ciclofosfamida/toxicidad , Estudios de Factibilidad , Humanos , Inyecciones Espinales , Macaca mulatta , Neoplasias Meníngeas/tratamiento farmacológico , Neoplasias Meníngeas/metabolismo , Meningitis/tratamiento farmacológico , Meningitis/metabolismo , Neoplasias , Resultado del Tratamiento
7.
J Clin Oncol ; 23(3): 525-31, 2005 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-15659498

RESUMEN

PURPOSE: A phase I trial of intrathecal (IT) mafosfamide was performed to determine the optimal dose, dose-limiting toxicities, and incidence and severity of other toxicities when administered in association with concomitant multiagent systemic chemotherapy to children younger than 3 years with newly diagnosed embryonal tumors. PATIENTS AND METHODS: Twenty-five assessable patients received IT mafosfamide at one of six dose levels ranging from 5 mg to 17 mg. Patients were premedicated with dexamethasone (0.15 mg/kg) and morphine (0.1 mg/kg) before receiving IT mafosfamide. Serial samples of CSF for pharmacokinetic studies were obtained in a subset of patients with Ommaya reservoirs. RESULTS: Irritability, presumably secondary to pain or headache during mafosfamide administration, was dose limiting in two of three patients at the 17-mg dose level. The maximum-tolerated dose of IT mafosfamide following premedication with dexamethasone and morphine was 14 mg. CONCLUSION: The maximum tolerated dose and recommended phase II dose of IT mafosfamide in patients younger than 3 years with newly diagnosed embryonal CNS tumors is 14 mg. A trial to assess the efficacy of regional therapy with IT mafosfamide administered with intensive systemic chemotherapy in children younger than 3 years with primary intracranial embryonal tumors is now in progress.


Asunto(s)
Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Ciclofosfamida/análogos & derivados , Ciclofosfamida/uso terapéutico , Neoplasias de Células Germinales y Embrionarias/tratamiento farmacológico , Afecto , Factores de Edad , Analgésicos Opioides/administración & dosificación , Antineoplásicos/administración & dosificación , Neoplasias Encefálicas/patología , Preescolar , Ciclofosfamida/administración & dosificación , Ciclofosfamida/efectos adversos , Dexametasona/administración & dosificación , Femenino , Glucocorticoides/administración & dosificación , Cefalea/inducido químicamente , Humanos , Lactante , Inyecciones Espinales , Masculino , Dosis Máxima Tolerada , Morfina/administración & dosificación , Neoplasias de Células Germinales y Embrionarias/patología , Dolor/etiología
8.
J Clin Oncol ; 20(1): 88-95, 2002 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11773158

RESUMEN

PURPOSE: We conducted a phase I trial of escalating doses of topotecan (TOPO) in association with a fixed systemic exposure of carboplatin (CARBO) with or without granulocyte colony-stimulating factor (G-CSF) in children. PATIENTS AND METHODS: Two separate cohorts of patients (pts) with solid tumors were studied: (A) pts with refractory or recurrent disease and (B) pts with no prior myelosuppressive therapy or newly diagnosed tumors for which there was no standard chemotherapy. CARBO was given on day 1 at an area under the curve of 6.5, followed by TOPO as a continuous infusion for 3 days; the starting dose of TOPO was 0.50 mg/m(2)/d. Cycles were repeated every 21 days. G-CSF was given at a dose of 5 microg/kg/d starting on day 4. RESULTS: Forty-eight of 51 pts were assessable for toxicity. In group A, dose-limiting myelosuppression persisted despite de-escalation of TOPO to 0.3 mg/m(2)/d and use of G-CSF. In group B, the maximum-tolerated dose of TOPO was 0.5 mg/m(2)/d for 3 days, and 0.6 mg/m(2)/d for 3 days with G-CSF. No significant nonhematologic toxicities were observed. Among 46 pts assessable for response, one had complete response, five had partial response, and 18 had stable disease. CONCLUSION: Although this combination possesses antineoplastic activity in pediatric solid tumors, hematologic toxicity precluded any meaningful TOPO dose escalation. The addition of G-CSF did not alter this. The potential for preservation of activity and diminution of toxicity with alternative sequences and schedules of administration (topoisomerase followed by alkylating or platinating agents) should be evaluated.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias/tratamiento farmacológico , Terapia Recuperativa/métodos , Topotecan/farmacología , Adolescente , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Carboplatino/administración & dosificación , Carboplatino/farmacocinética , Niño , Preescolar , Femenino , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Humanos , Lactante , Funciones de Verosimilitud , Masculino , Dosis Máxima Tolerada , Topotecan/administración & dosificación , Topotecan/farmacocinética
9.
J Clin Oncol ; 21(1): 143-7, 2003 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-12506183

RESUMEN

PURPOSE: A phase I trial of intrathecal (IT) topotecan was performed to determine the optimal dose, the dose-limiting toxic effects, and the incidence and severity of other toxic effects in patients 3 years and older with neoplastic meningitis. PATIENTS AND METHODS: Twenty-three assessable patients received IT topotecan administered by means of either lumbar puncture or an indwelling ventricular access device (Ommaya reservoir). Intrapatient dose escalation from 0.025 mg to 0.2 mg was performed in the first cohort of patients. Subsequent cohorts of patients were treated at fixed dose levels of 0.2 mg, 0.4 mg, or 0.7 mg. Serial samples of CSF for pharmacokinetic studies were obtained in a subset of patients with Ommaya reservoirs. RESULTS: Arachnoiditis, characterized by fever, nausea, vomiting, headache, and back pain, was the dose-limiting toxic effect in two of four patients enrolled at the 0.7 mg dose level. The maximum-tolerated dose (MTD) was 0.4 mg. Six of the 23 assessable patients had evidence of benefit manifested as prolonged disease stabilization or response. CONCLUSION: The MTD and recommended phase II dose of IT topotecan in patients who are 3 years or older is 0.4 mg. A phase II trial of IT topotecan in children with neoplastic meningitis is in progress.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Meníngeas/tratamiento farmacológico , Topotecan/uso terapéutico , Adolescente , Adulto , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Niño , Preescolar , Femenino , Humanos , Inyecciones Espinales , Masculino , Persona de Mediana Edad , Topotecan/efectos adversos , Topotecan/farmacocinética
10.
Clin Cancer Res ; 8(7): 2202-9, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12114421

RESUMEN

PURPOSE: The purpose of this study was to determine the effect of enzyme-inducing anticonvulsants (EIAs) on the disposition of irinotecan and metabolites in pediatric patients with high-grade glioma. EXPERIMENTAL DESIGN: Pediatric patients with newly diagnosed high-grade glioma were enrolled on this study between March 1999 and February 2001. During course 1, irinotecan was administered as a 60-min i.v. infusion at a dosage of 20 mg/m(2)/day for 5 days of 2 consecutive weeks. On days 1 and 12 of course 1, we collected serial plasma samples to measure the concentrations of the lactone and total forms of irinotecan and its metabolites SN-38 (7-ethyl-10-hydroxycamptothecin), SN-38 glucuronide (7-ethyl-10-[3,4,5-trihydroxy-pyran-2-carboxylic acid]camptothecin), and 7-ethyl-10-[4-N-(5-aminopentanoic acid)-1-piperidino]carbonyloxycamptothecin. RESULTS: Thirty-one patients were enrolled. In patients receiving EIAs, the area under the concentration versus time curve (AUC) of irinotecan lactone and SN-38 lactone was significantly lower (P = 0.01 and P = 0.002, respectively), and the irinotecan lactone clearance was significantly higher (P = 0.0003), as compared with those in patients who received no EIAs. The glucuronidation ratio was higher (P = 0.0009), and the ratio of SN-38 AUC to irinotecan AUC was lower (P = 0.02) in patients who received EIAs. Two patients receiving EIAs tolerated increased irinotecan dosages of 30 and 40 mg/m(2)/day without toxicity. One patient receiving EIAs experienced grade 3 diarrhea when the dosage of irinotecan was increased to 60 mg/m(2)/day. CONCLUSIONS: EIAs increase the clearance of irinotecan and cause a decrease in systemic exposure to the active metabolite SN-38. Patients who are receiving irinotecan and who require anticonvulsants should be placed on non-EIA therapy, when possible.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Antineoplásicos Fitogénicos/farmacocinética , Neoplasias Encefálicas/metabolismo , Camptotecina/análogos & derivados , Camptotecina/farmacocinética , Inhibidores Enzimáticos/farmacocinética , Glioma/metabolismo , Inhibidores de Topoisomerasa I , Adolescente , Adulto , Neoplasias Encefálicas/tratamiento farmacológico , Niño , Preescolar , Relación Dosis-Respuesta a Droga , Inducción Enzimática , Femenino , Glioma/tratamiento farmacológico , Humanos , Infusiones Intravenosas , Irinotecán , Masculino
11.
Neuro Oncol ; 5(4): 261-7, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14565163

RESUMEN

Idarubicin (IDA), the 4-demethoxy analog of daunomycin, has had significant cytotoxicity in many malignancies. In previous reports, the alcohol metabolite of IDA, 4-demethoxydaunorubicinol (idarubicinol, or IDOL), had cytotoxic activity and the ability to penetrate the blood-brain barrier. For this reason, the Pediatric Oncology Group conducted a Phase 2 trial of IDA for children with recurrent or progressive brain tumors. Ninety-one eligible children were entered on this study, with ages ranging from 3 months to 19 years. Patients were stratified by tumor types into 6 categories: stratum 1, low-grade astrocytoma; stratum 2, malignant glioma (glioblastoma multiforme and anaplastic astrocytoma); stratum 3, medulloblastoma; stratum 4, brainstem glioma; stratum 5, ependymoma; and stratum 6, miscellaneous malignant tumors not included in the previous diagnoses. IDA(18 mg/m2) was infused over 4 h and followed by granulocyte colony-stimulating factor (G-CSF) beginning day 5 after infusion of IDA. G-CSF was continued until blood cell count recovery. Cycles were repeated at approximately 21-day intervals until patients developed progressive disease or had completed 6 cycles with stable or improved disease. Pharmacokinetic plasma and cerebrospinal fluid (CSF) samples were collected from a subset of these patients. Response was poor in all strata. Most patients developed progressive disease; however, in 21 patients with medulloblastoma there was 1 partial response, and 6 patients had stable disease (SD) that in 4 patients lasted more than 20 weeks. Grades 3/4 hematopoietic toxicities were the most common toxic events, and 14 infection-related events resulted in hospitalization of patients. Only 1 patient developed reduced cardiac function. The systemic clearance data for IDA and IDOL were nearly identical to those published on patients with leukemia, and the plasma elimination of the IDOL metabolite was substantially longer than that of the parent drug IDA. The peak CSF:plasma ratios of IDA and IDOL were very low. The overall response rates to IDA were disappointing despite periods of prolonged SD in nearly a fourth of the patients. We conclude from this data and from that in nonhuman primates that it is unlikely that IDA, daunomycin, or other related anthracyclines will be useful for treating primary CNS tumors.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Idarrubicina/uso terapéutico , Adolescente , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Niño , Preescolar , Femenino , Humanos , Idarrubicina/farmacocinética , Lactante , Masculino
12.
Int J Radiat Oncol Biol Phys ; 52(2): 325-32, 2002 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11872277

RESUMEN

PURPOSE: To estimate the local control and patterns of failure for pediatric patients with low-grade astroglial tumors (LGA) and ependymoma (EP) treated with three-dimensional conformal radiation therapy (CRT) using an anatomically defined clinical target volume (CTV). METHODS AND MATERIALS: From an ongoing, prospective Phase II trial initiated in July 1997, 102 pediatric patients with LGA (n = 38) and EP (n = 64) have been treated with CRT using an anatomically defined CTV extending 1.0 cm beyond the gross tumor volume and a 0.5-cm margin (planning target volume) extending outside of the CTV. The prescribed dose was 54 Gy (LGA) and 59.4 Gy (EP). RESULTS: Patients with EP have been followed for a median of 17 months (range 3--43 months), and six failures have occurred. Patients with LGA have been followed for a median of 17 months (3--44 months), and four failures have occurred. Three-dimensional magnetic resonance (MR) studies performed to document treatment failure were registered with the MR and computed tomography (CT) data used in the treatment planning process. Failure occurred within the CTV for 5 patients with EP, including 3 with concurrent subarachnoid dissemination. One patient with EP developed metastatic disease with no evidence of local failure. Three patients with LGA failed within the CTV and one failed immediately outside of the CTV. CONCLUSIONS: Treatment of an anatomically defined CTV, encompassing 1.0 cm of non-involved brain beyond the margin of resection or neuroimaging-defined tumor, appears to be safe for pediatric patients with LGA and EP based on these preliminary data. Normal tissue sparing through the use of advanced radiation therapy treatment planning and delivery techniques should be beneficial to pediatric patients if the rate and patterns of failure are similar to conventional techniques and toxicity reduction can be objectively documented.


Asunto(s)
Astrocitoma/radioterapia , Neoplasias Encefálicas/radioterapia , Ependimoma/radioterapia , Radioterapia Conformacional/métodos , Adolescente , Astrocitoma/patología , Neoplasias Encefálicas/patología , Niño , Preescolar , Ependimoma/patología , Femenino , Estudios de Seguimiento , Humanos , Masculino , Estudios Prospectivos , Planificación de la Radioterapia Asistida por Computador
13.
Int J Radiat Oncol Biol Phys ; 53(1): 52-7, 2002 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-12007941

RESUMEN

PURPOSE: To investigate the influence of histologic grade on progression-free survival (PFS) after irradiation (RT) for pediatric patients with localized ependymoma. METHODS AND MATERIALS: Fifty patients with localized ependymoma (median age 3.6 years, range 1-18 years at the time of RT) were treated with RT between December 1982 and June 1999. Anaplastic features were identified in 14 of 50 patients. The extent of resection was characterized as gross-total in 36 patients, near-total in 5, and subtotal in 9. The median dose to the primary site was 54 Gy. Of the 50 patients, 23 received pre-RT chemotherapy. RESULTS: Thirty-nine patients were alive at a median follow-up of 46 months (range 21-214) from diagnosis. Thirty-four patients remained progression free at a median follow-up of 35 months (range 13-183) after the initiation of RT. Progression occurred in 16 patients (12 local and 4 local and distant), with a median time to failure of 21.2 months (range 4.6-65.0). The tumor grade significantly influenced the PFS after RT (p < 0.0005). The estimated 3-year PFS rate was 28% +/- 14% for patients with anaplastic ependymoma compared with 84% +/- 8% for patients with differentiated ependymoma. These results remained significant when corrected for age at diagnosis (<3 years), pre-RT chemotherapy, and extent of resection. Patients who received pre-RT chemotherapy had an inferior 3-year PFS estimate after RT (49 +/- 12%) compared with those who did not (84% +/- 10%; p = 0.056). Anaplastic ependymoma was found more frequently in the supratentorial brain (p = 0.002). Six of 12 patients with supratentorial tumor developed recurrence; recurrence was restricted to patients with anaplastic ependymoma. CONCLUSION: Tumor grade influences outcome for patients with ependymoma independent of other factors and should be considered in the design and analysis of prospective trials involving pediatric patients treated with RT. Chemotherapy before RT influences the PFS and overall survival after RT. The effect is more pronounced when progression occurs during chemotherapy.


Asunto(s)
Neoplasias Encefálicas/patología , Neoplasias Encefálicas/radioterapia , Ependimoma/patología , Ependimoma/radioterapia , Adolescente , Neoplasias Encefálicas/tratamiento farmacológico , Niño , Preescolar , Terapia Combinada , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Ependimoma/tratamiento farmacológico , Femenino , Humanos , Lactante , Masculino , Dosificación Radioterapéutica , Estudios Retrospectivos , Neoplasias Supratentoriales/tratamiento farmacológico , Neoplasias Supratentoriales/patología , Neoplasias Supratentoriales/radioterapia , Resultado del Tratamiento
14.
Int J Radiat Oncol Biol Phys ; 58(4): 1194-207, 2004 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15001264

RESUMEN

PURPOSE: Central nervous system (CNS) irradiation can cause sensorineural hearing loss. The relationship between the dose to the cochlea and the development of hearing loss is unknown. Conformal radiation therapy (CRT) techniques facilitate accurate cochlear dosimetry. We modeled hearing threshold levels (HTL) after CRT in children with localized primary brain tumors (ependymoma, low- or high-grade astrocytoma, craniopharyngioma, or CNS germinoma) by using cochlear dose and clinical variables. PATIENTS AND METHODS: We evaluated 72 children (median age, 9.5 years) with audiograms before and every 6 months after CRT (median follow-up, 16.6 months; range, 4.3-42.6 months). We used a mixed-effects model to predict change in hearing for each ear as a function of time, cochlear dose, and clinical variables. RESULTS: Hearing was affected the greatest in patients with CSF shunts and pre-CRT ototoxic chemotherapy, enhanced by cochlear dose, and was more prominent on the right side. Hearing impairment after CRT alone occurred at low and intermediate frequencies in patients with shunts and supratentorial tumors when the cochlear dose exceeded 32 Gy. Patients with shunts and central supratentorial tumors developed intermediate-frequency hearing loss after CRT alone regardless of dose. CONCLUSIONS: Hearing loss during the first 4 years after CRT alone is uncommon, although patients with shunts and supratentorial tumors appear to be at increased risk for low- and intermediate-frequency effects. CSF shunting and increased cochlear dose enhance the effect of ototoxic chemotherapy. If possible, the average cochlear dose should be <32 Gy over a 6-week course of treatment until more specific dose data become available.


Asunto(s)
Neoplasias Encefálicas/radioterapia , Cóclea/efectos de la radiación , Irradiación Craneana/efectos adversos , Pérdida Auditiva/etiología , Radioterapia Conformacional/efectos adversos , Adolescente , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Audiometría , Neoplasias Encefálicas/tratamiento farmacológico , Niño , Preescolar , Femenino , Estudios de Seguimiento , Pérdida Auditiva/patología , Pérdida Auditiva Sensorineural/etiología , Humanos , Masculino , Dosificación Radioterapéutica
15.
Cancer Chemother Pharmacol ; 52(6): 435-41, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-13680158

RESUMEN

PURPOSE: To construct a population pharmacokinetic model for temozolomide (TMZ), a novel imidazo-tetrazine methylating agent and its metabolites MTIC and AIC in infants and children with primary central nervous system tumors. METHODS: We evaluated the pharmacokinetics of TMZ and MTIC in 39 children (20 boys and 19 girls) with 132 pharmacokinetic studies (109 in the training set and 23 in the validation set). The median age was 7.1 years (range 0.7 to 21.9 years). Children received oral TMZ dosages ranging from 145 to 200 mg/m(2) per day for 5 days in each course of therapy. Serial plasma samples were collected after the first and fifth doses of the first and third courses. Approximately eight plasma samples were collected up to 8 h after each dose, and assayed for TMZ, MTIC, and AIC by HPLC with UV detection. A one-compartment model was fitted to the TMZ and metabolite plasma concentrations using maximum likelihood estimation. Covariates, including demographics and biochemical data were tested for their effects on TMZ clearance (CL/F) and MTIC AUC utilizing a two-stage approach via linear mixed-effects modeling. RESULTS: The population mean (inter- and intrapatient variability expressed as %CV) for the pharmacokinetic parameters (based on the training set) were as follows: TMZ CL/F 5.4 l/h (53.4, 17.5), Vc/F 14.0 l (48.5, 39.2), C(max) 9.1 mg/l (20.8, 29.1), and MTIC AUC 1.0 microg/ml.h (13.9, 30.0). Covariate analysis showed that increasing age and body surface area (BSA) were associated with a significant increases in TMZ CL, Vc, and C(max) ( P<0.05), and that increasing age was associated with significant decreases in TMZ and MTIC AUC. Indicators of liver and renal function were not significantly associated with TMZ pharmacokinetics or MTIC AUC. The final model with the significant covariates was validated using the remaining 23 pharmacokinetic studies. CONCLUSIONS: This study extends previous work done in adults, and identified BSA and age as covariates that account for variability in TMZ disposition in infants and children with primary CNS malignancies.


Asunto(s)
Antineoplásicos Alquilantes , Neoplasias Encefálicas/metabolismo , Dacarbazina , Dacarbazina/análogos & derivados , Adolescente , Adulto , Factores de Edad , Antineoplásicos Alquilantes/sangre , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacocinética , Área Bajo la Curva , Disponibilidad Biológica , Superficie Corporal , Niño , Preescolar , Cromatografía Líquida de Alta Presión , Dacarbazina/sangre , Dacarbazina/metabolismo , Dacarbazina/farmacocinética , Femenino , Humanos , Lactante , Modelos Lineales , Masculino , Tasa de Depuración Metabólica , Reproducibilidad de los Resultados , Temozolomida
16.
Neuro Oncol ; 13(4): 410-6, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21345842

RESUMEN

An open-label phase II study (ACNS0126) testing the efficacy of chemoradiotherapy with temozolomide (TMZ) followed by adjuvant TMZ was conducted by the Children's Oncology Group. During the period from July 6, 2004 through September 6, 2005, 63 children with newly diagnosed diffuse intrinsic pontine glioma (DIPG) were enrolled in the study. All patients received TMZ at a dosage of 90 mg/m(2)/day for 42 days to a dose of 59.4 Gy. Four weeks following irradiation, TMZ was given at a dosage of 200 mg/m(2)/day for 5 days every 28 days, for a total of 10 cycles. The primary objective of the statistical analysis was to determine whether the current treatment produced a 1-year event-free survival (EFS) rate higher than the historical baseline of 21.9% observed in CCG-9941. The mean 1-year EFS (± standard deviation) was 14% ± 4.5%, compared with 21.9% ± 5% for CCG-9941. The P value of the test of comparison of 1-year EFS, based on a 1-sided, 1-sample test of proportions, was .96. There was no evidence that temozolomide produced a 1-year EFS rate higher than 21.9%. The mean 1-year OS (± standard deviation) was 40% ± 6.5%, compared with 32% ± 6% for CCG-9941. The median time to death was 9.6 months. Chemoradiotherapy with TMZ followed by adjuvant TMZ is not more effective than previously reported regimens for the treatment of children with DIPG.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Neoplasias del Tronco Encefálico/tratamiento farmacológico , Dacarbazina/análogos & derivados , Puente , Adolescente , Neoplasias del Tronco Encefálico/radioterapia , Quimioterapia Adyuvante , Niño , Preescolar , Terapia Combinada , Dacarbazina/uso terapéutico , Femenino , Humanos , Masculino , Dosificación Radioterapéutica , Tasa de Supervivencia , Temozolomida , Resultado del Tratamiento
17.
Brain Pathol ; 21(3): 349-50, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21492294

RESUMEN

Pediatric meningiomas are rare and account for about 1.5% of all intracranial tumors. When compared to adults, intraventricular location of childhood meningiomas is four to ten times as high. Atypical pathology of these lesions is very uncommon and indicates an aggressive nature. They are usually associated with Neurofibromatosis 2 (NF2) or previous cranial irradiation. Here, we present an interesting case of an unusually large, congenital intraventricular meningioma of atypical pathology in a 16 month old child with subsequently diagnosed NF2. A brief review of literature is also presented with this case illustration.


Asunto(s)
Neoplasias del Ventrículo Cerebral/patología , Meningioma/patología , Neurofibromatosis 2/diagnóstico , Neoplasias del Ventrículo Cerebral/cirugía , Análisis Mutacional de ADN , Femenino , Genes de la Neurofibromatosis 2 , Humanos , Lactante , Meningioma/cirugía , Neurofibromatosis 2/genética , Neurofibromatosis 2/patología , Neurofibromatosis 2/cirugía
18.
Neuro Oncol ; 13(3): 317-23, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21339192

RESUMEN

To determine whether temozolomide is an active agent in the treatment of children with high-grade astrocytomas and whether survival is influenced by the expression of the O6-methylguanine-methyltransferase gene (MGMT) in these patients. In the Children's Oncology Group study ACNS0126, 107 patients with a diagnosis of anaplastic astrocytoma (AA), glioblastoma multiforme (GBM), or gliosarcoma were enrolled. All patients underwent concomitant chemoradiotherapy with temozolomide, followed by adjuvant chemotherapy with temozolomide. The outcomes were compared with those of children treated in Children's Cancer Group (CCG) study CCG-945. Formalin-fixed, paraffin-embedded tumor tissue was available in 71 cases for immunohistochemical analysis of MGMT expression. Ninety patients were deemed eligible, 31 with AA, 55 with GBM, and 4 with other eligible diagnoses. The 3-year event-free survival (EFS) and overall survival (OS) rates were 11 ± 3% and 22 ± 5%, respectively. There was no evidence that temozolomide given during radiation therapy and as adjuvant therapy resulted in improved EFS compared with that found in CCG-945 (p = 0.98). The 3-year EFS rate for AA was 13 ± 6% in ACNS0126 compared with 22 ± 5.5% in CCG-945 (p = 0.95). The 3-year EFS rate for GBM was 7 ± 4% in ACNS0126 compared with 15 ± 5% in CCG-945 (p = 0.77). The 2-year EFS rate was 17 ± 5% among patients without MGMT overexpression and 5 ± 4% among those with MGMT overexpression (p = 0.045). Temozolomide failed to improve outcome in children with high-grade astrocytomas. MGMT overexpression was adversely associated with survival.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Dacarbazina/análogos & derivados , Glioblastoma/tratamiento farmacológico , Gliosarcoma/tratamiento farmacológico , Adolescente , Adulto , Neoplasias Encefálicas/radioterapia , Quimioterapia Adyuvante , Niño , Preescolar , Terapia Combinada , Dacarbazina/uso terapéutico , Femenino , Estudios de Seguimiento , Glioblastoma/radioterapia , Gliosarcoma/radioterapia , Humanos , Técnicas para Inmunoenzimas , Masculino , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Dosificación Radioterapéutica , Tasa de Supervivencia , Temozolomida , Resultado del Tratamiento , Adulto Joven
20.
Pediatr Blood Cancer ; 47(3): 293-8, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16206208

RESUMEN

BACKGROUND: Juvenile pilocytic astrocytomas (JPAs) are the most common central nervous system tumors in children. If completely resected, JPAs are associated with an excellent outcome. However, there is need for additional therapeutic approaches for those JPAs which are incompletely resected and fail subsequent standard chemotherapy/radiation. To explore the possibility for a novel therapeutic approach we measured the effect of the epidermal growth factor receptor (EGFR) small molecule tyrosine kinase inhibitor gefitinib on five JPA primary cell-cultures. PROCEDURE: Due to a lack of established cell-lines of JPA very few in vitro drug sensitivity assays have been performed. In this study we have succeeded in propagating short-term primary cell-cultures established from surgical specimens. The effect of gefitinib on proliferation in JPA derived primary cell-cultures was measured by a standard tritiated thymidine incorporation assay. The level of expression of EGFR, the intended target of gefitinib, was measured by immunohistochemistry, flow cytometry and RT-PCR. RESULTS: Gefitinib was shown to inhibit proliferation in all five JPA cell-cultures tested, with IC-50's between 1.6 and 9.6 microM. However, EGFR protein and mRNA expression was undetectable. Further studies with cetuximab, an EGFR-specific inhibitory monoclonal antibody, showed no effect on proliferation in JPA. CONCLUSIONS: Based on these preclinical data, gefitinib may be a suitable salvage chemotherapy drug to explore further in those patients with JPA who have recurred after primary chemotherapy. Of interest, it appears that the anti-tumor effect of gefitinib in JPA cell-cultures may be mediated through a pathway other than EGFR inhibition.


Asunto(s)
Astrocitoma/tratamiento farmacológico , Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Quinazolinas/farmacología , Astrocitoma/metabolismo , Astrocitoma/patología , Proliferación Celular/efectos de los fármacos , Neoplasias del Sistema Nervioso Central/metabolismo , Neoplasias del Sistema Nervioso Central/patología , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Receptores ErbB/genética , Receptores ErbB/metabolismo , Citometría de Flujo , Gefitinib , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sensibilidad y Especificidad , Timidina/metabolismo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA