Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Pediatr Blood Cancer ; 70(7): e30365, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37073741

RESUMEN

BACKGROUND: Survival for children with metastatic hepatoblastoma (HB) remains suboptimal. We report the response rate and outcome of two courses of vincristine/irinotecan/temsirolimus (VIT) in children with high-risk (HR)/metastatic HB. PROCEDURES: Patients with newly diagnosed HB received HR window chemotherapy if they had metastatic disease or a serum alpha-fetoprotein (AFP) level less than 100 ng/mL. Patients received vincristine (days 1 and 8), irinotecan (days 1-5), and temsirolimus (days 1 and 8). Cycles were repeated every 21 days. Responders had either a 30% decrease using RECIST (Response Evaluation Criteria in Solid Tumors) criteria OR a 90% (>1 log10 decline) AFP decline after two cycles. Responders received two additional cycles of VIT intermixed with six cycles of cisplatin/doxorubicin/5-fluorouracil/vincristine (C5VD). Nonresponders received six cycles of C5VD alone. RESULTS: Thirty-six eligible patients enrolled on study. The median age at enrollment was 27 months (range: 7-170). Seventeen of 36 patients were responders (RECIST and AFP = 3, RECIST only = 4, AFP only = 10). The median AFP at diagnosis was 222,648 ng/mL and the median AFP following two VIT cycles was 19,262 ng/mL. Three-year event-free survival was 47% (95% confidence interval [CI]: 30%-62%), while overall survival was 67% (95% CI: 49%-80%). CONCLUSION: VIT did not achieve the study efficacy endpoint. Temsirolimus does not improve the response rate seen in patients treated with vincristine and irinotecan (VI) alone as part of the initial treatment regimen explored in this study. Additionally, AFP response may be a more sensitive predictor of disease response than RECIST in HB.


Asunto(s)
Hepatoblastoma , Neoplasias Hepáticas , Niño , Humanos , Hepatoblastoma/patología , Irinotecán/uso terapéutico , Vincristina , Neoplasias Hepáticas/patología , alfa-Fetoproteínas , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Resultado del Tratamiento
2.
Cancer ; 128(5): 1057-1065, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34762296

RESUMEN

BACKGROUND: The Children's Oncology Group (COG) adopted cisplatin, 5-flourouracil, and vincristine (C5V) as standard therapy after the INT-0098 legacy study showed statistically equivalent survival but less toxicity in comparison with cisplatin and doxorubicin. Subsequent experience demonstrated doxorubicin to be effective in patients with recurrent disease after C5V, and this suggested that it could be incorporated to intensify therapy for patients with advanced disease. METHODS: In this nonrandomized, phase 3 COG trial, the primary aim was to explore the feasibility and toxicity of a novel therapeutic cisplatin, 5-flourouracil, vincristine, and doxorubicin (C5VD) regimen with the addition of doxorubicin to C5V for patients considered to be at intermediate risk. Patients were eligible if they had unresectable, nonmetastatic disease. Patients with a complete resection at diagnosis and local pathologic evidence of small cell undifferentiated histology were also eligible for an assessment of feasibility. RESULTS: One hundred two evaluable patients enrolled between September 14, 2009, and March 12, 2012. Delivery of C5VD was feasible and tolerable: the mean percentages of the target doses delivered were 96% (95% CI, 94%-97%) for cisplatin, 96% (95% CI, 94%-97%) for 5-fluorouracil, 95% (95% CI, 93%-97%) for doxorubicin, and 90% (95% CI, 87%-93%) for vincristine. Toxicity was within expectations, with death as a first event in 1 patient. The most common adverse events were febrile neutropenia (n = 55 [54%]), infection (n = 48 [47%]), mucositis (n = 31 [30%]), hypokalemia (n = 39 [38%]), and elevated aspartate aminotransferase (n = 28 [27%]). The 5-year event-free and overall survival rates for the 93 patients who did not have complete resection at diagnosis were 88% (95% CI, 79%-93%) and 95% (95% CI, 87%-98%), respectively. CONCLUSIONS: The addition of doxorubicin to the previous standard regimen of C5V is feasible, tolerable, and efficacious, and this suggests that C5VD is a good regimen for future clinical trials.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Hepatoblastoma , Neoplasias Hepáticas , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Cisplatino/efectos adversos , Doxorrubicina/efectos adversos , Estudios de Factibilidad , Hepatoblastoma/tratamiento farmacológico , Hepatoblastoma/patología , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Resultado del Tratamiento , Vincristina/efectos adversos
3.
Clin Pharmacokinet ; 58(7): 899-910, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30810947

RESUMEN

BACKGROUND: Infants with acute lymphoblastic leukemia (ALL) treated with high-dose methotrexate may have reduced methotrexate clearance (CL) due to renal immaturity, which may predispose them to toxicity. OBJECTIVE: The aim of this study was to develop a population pharmacokinetic (PK) model of methotrexate in infants with ALL. METHODS: A total of 672 methotrexate plasma concentrations were obtained from 71 infants enrolled in the Children's Oncology Group (COG) Clinical Trial P9407. Infants received methotrexate 4 g/m2 intravenously for four cycles during weeks 4-12 of intensification. A population PK analysis was performed using NONMEM® version 7.4. The final model was evaluated using a non-parametric bootstrap and a visual predictive check. Simulations were performed to evaluate methotrexate dose and the utility of a bedside algorithm for dose individualization. RESULTS: Methotrexate was best characterized by a two-compartment model with allometric scaling. Weight was the only covariate included in the final model. The coefficient of variation for interoccasion variability (IOV) on CL was relatively high at 25.4%, compared with the interindividual variability for CL and central volume of distribution (10.7% and 13.2%, respectively). Simulations identified that 21.1% of simulated infants benefitted from bedside dose adjustment, and adjustment of methotrexate doses during infusions can avoid supratherapeutic concentrations. CONCLUSION: Infants treated with high-dose methotrexate demonstrated a relatively high degree of IOV in methotrexate CL. The magnitude of IOV in the CL of methotrexate suggests that use of a bedside algorithm may avoid supratherapeutic methotrexate concentrations resulting from high IOV in methotrexate CL.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacocinética , Metotrexato/farmacocinética , Modelos Biológicos , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Femenino , Humanos , Lactante , Masculino , Medicina de Precisión
4.
Cancer Chemother Pharmacol ; 83(2): 349-360, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30488179

RESUMEN

PURPOSE: High-dose methotrexate (HDMTX) is critical to the successful treatment of pediatric acute lymphoblastic leukemia (ALL) but can cause significant toxicities. This study prospectively evaluated the effectiveness of a fixed algorithm which requires no real-time pharmacokinetic modeling and no previous patient exposure to HDMTX, to individualize HDMTX dosing for at-risk patients with the aim of avoiding methotrexate-related toxicities. METHODS: We developed a simple algorithm to individualize HDMTX infusions with 0-2 rate adjustments based on methotrexate levels during the infusion. This was a prospective, open-label, study; eligible patients were identified and referred by their oncologist. RESULTS: Fifty-four evaluable cycles of HDMTX (5 g/m2 over 24 h) were administered to 22 patients. Blood samples were obtained in 21 patients to examine single nucleotide polymorphisms (SNPs) related to methotrexate disposition. Twelve (54.5%) subjects had a history of previous HDMTX toxicities including seven (31.8%) who previously required glucarpidase rescue and seven (31.8%) with an entry glomerular filtration rate < 80 ml/min/1.73 m2. 107/110 (97.2%) of methotrexate levels were drawn properly and 100% of algorithm dosing instructions were performed correctly at the bedside. Thirty-five (64.8%) of all cycles and 24 of 33 (72.7%) cycles that required a dose-adjustment had an end 24-h methotrexate level (Cpss) within our goal range of 65 ± 15 µM with only 3 (5.6%) resulting in Cpss higher than goal. Grade 3/4 toxicities were rare; no patients developed > Grade 1 acute kidney injury. CONCLUSION: This algorithm is a simple, safe and effective method for individualizing HDMTX in pediatric patients with ALL. CLINICALTRIALS. GOV REGISTRY: NCT02076997.


Asunto(s)
Algoritmos , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/farmacocinética , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Metotrexato/administración & dosificación , Metotrexato/farmacocinética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Adolescente , Adulto , Niño , Preescolar , Relación Dosis-Respuesta a Droga , Femenino , Estudios de Seguimiento , Humanos , Lactante , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Pronóstico , Estudios Prospectivos , Factores de Riesgo , Distribución Tisular , Adulto Joven
5.
J Pediatr Hematol Oncol ; 39(1): 72-76, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27820134

RESUMEN

We developed a bedside algorithm for individually adjusting the high-dose methotrexate (HDMTX) dose (5 g/m) given to patients with acute lymphoblastic leukemia at high risk for methotrexate toxicity. Data were reviewed for 8 patients receiving 21 cycles of HDMTX as per our algorithm. Eleven cycles began with 5 g/m, 10 cycles began with a preinfusion 20% to 25% dose reduction. Neither mean MTX AUC (2320.5±179.1 vs. 2080.4±161.7 µmol×h/L), mean Cpss (64.3±7.9 vs. 60.8±6.1 µM), nor toxicities were statistically different between groups. Our algorithm allowed the safe administration of HDMTX to patients at risk of MTX toxicities and obviated the need for preinfusion dose reduction.


Asunto(s)
Algoritmos , Metotrexato/administración & dosificación , Sistemas de Atención de Punto , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/prevención & control , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Área Bajo la Curva , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Monitoreo de Drogas , Fluidoterapia , Humanos , Infusiones Intravenosas , Metotrexato/efectos adversos , Metotrexato/farmacocinética , Mucositis/inducido químicamente , Mucositis/prevención & control , Enfermedades del Sistema Nervioso/inducido químicamente , Enfermedades del Sistema Nervioso/prevención & control , Estudios Retrospectivos
6.
Cancer Chemother Pharmacol ; 79(1): 181-187, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28032129

RESUMEN

PURPOSE: Characterize the pharmacokinetics of oral crizotinib in children with cancer. METHODS: Sixty-four children with solid tumors or anaplastic large-cell lymphoma (ALCL) enrolled on a phase 1/2 trial of the ALK, MET and ROS1 inhibitor, crizotinib, had pharmacokinetic sampling after the first dose (n = 15) or at steady state (n = 49). Dose levels studied were 100, 130, 165, 215, 280 and 365 mg/m2/dose administered twice daily. Two capsule and two oral liquid formulations were used over the course of the trial. Crizotinib was quantified with a validated HPLC/tandem mass spectrometry method with a lower limit of detection of 0.2 ng/mL. Pharmacokinetic parameters were derived using non-compartmental analysis. RESULTS: Time to peak plasma concentration was 4 h. At 280 mg/m2 (MTD), mean (±SD) steady-state peak plasma concentration was 717 ± 201 ng/mL, and steady-state trough plasma concentration was 480 ± 176 ng/mL. At steady state, AUC0-τ was proportional to dose over the dose range of 215-365 mg/m2/dose. Apparent clearance of crizotinib was 731 ± 241 mL/min/m2. Steady-state AUC0-τ at 280 mg/m2/dose was 2.5-fold higher than the AUC0-∞ in adults receiving 250 mg (~140 mg/m2). Age, sex and drug formulation do not account for the inter-subject variability in AUC0-τ at steady state. The accumulation index was 4.9, and the half-life estimated from the accumulation index was 36 h. CONCLUSIONS: The pharmacokinetics of oral crizotinib in children is similar to that in adults. Steady-state trough-free crizotinib concentrations in plasma at the MTD exceed inhibitory concentrations of crizotinib in ALCL cell lines. CLINICALTRIALS. GOV IDENTIFIER: NCT00939770.


Asunto(s)
Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacocinética , Pirazoles/farmacocinética , Piridinas/farmacocinética , Administración Oral , Adolescente , Adulto , Niño , Preescolar , Crizotinib , Femenino , Humanos , Linfoma Anaplásico de Células Grandes/tratamiento farmacológico , Masculino , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirazoles/administración & dosificación , Piridinas/administración & dosificación
7.
J Hepatol ; 65(2): 325-33, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27117591

RESUMEN

BACKGROUND & AIMS: Pediatric liver cancer is a rare but serious disease whose incidence is rising, and for which the therapeutic options are limited. Development of more targeted, less toxic therapies is hindered by the lack of an experimental animal model that captures the heterogeneity and metastatic capability of these tumors. METHODS: Here we established an orthotopic engraftment technique to model a series of patient-derived tumor xenograft (PDTX) from pediatric liver cancers of all major histologic subtypes: hepatoblastoma, hepatocellular cancer and hepatocellular malignant neoplasm. We utilized standard (immuno) staining methods for histological characterization, RNA sequencing for gene expression profiling and genome sequencing for identification of druggable targets. We also adapted stem cell culturing techniques to derive two new pediatric cancer cell lines from the xenografted mice. RESULTS: The patient-derived tumor xenografts recapitulated the histologic, genetic, and biological characteristics-including the metastatic behavior-of the corresponding primary tumors. Furthermore, the gene expression profiles of the two new liver cancer cell lines closely resemble those of the primary tumors. Targeted therapy of PDTX from an aggressive hepatocellular malignant neoplasm with the MEK1 inhibitor trametinib and pan-class I PI3 kinase inhibitor NVP-BKM120 resulted in significant growth inhibition, thus confirming this PDTX model as a valuable tool to study tumor biology and patient-specific therapeutic responses. CONCLUSIONS: The novel metastatic xenograft model and the isogenic xenograft-derived cell lines described in this study provide reliable tools for developing mutation- and patient-specific therapies for pediatric liver cancer. LAY SUMMARY: Pediatric liver cancer is a rare but serious disease and no experimental animal model currently captures the complexity and metastatic capability of these tumors. We have established a novel animal model using human tumor tissue that recapitulates the genetic and biological characteristics of this cancer. We demonstrate that our patient-derived animal model, as well as two new cell lines, are useful tools for experimental therapies.


Asunto(s)
Neoplasias Hepáticas , Animales , Carcinoma Hepatocelular , Línea Celular Tumoral , Niño , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Ratones , Trasplante de Neoplasias , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Pediatr Surg ; 51(4): 525-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26607968

RESUMEN

BACKGROUND: In children diagnosed with hepatoblastoma (HB), the lungs are the most common site of metastasis at both initial presentation and relapse. Previous studies have encouraged pulmonary metastasectomy to achieve a disease-free state after resection of the primary hepatic lesion. However, there is no consensus about how to manage recurrent pulmonary metastasis. PROCEDURE: A retrospective, multi-institutional review was performed from 2005 to 2014 to identify HB patients ≤18years of age who had disease recurrence associated with pulmonary metastases alone. RESULTS: Ten patients between the ages of 8 and 33months were identified. Pulmonary metastatic recurrence was detected by measuring alpha-fetoprotein (AFP) levels and/or with CT scans of the chest. All patients subsequently underwent pulmonary metastasectomy without post-operative complications. At last follow-up, 8 patients were alive and had normal AFP levels. The 8 survivors had a median follow-up from therapy completion of 18.5months. Two patients who presented with extrapulmonary recurrence subsequently died of treatment refractory disease. CONCLUSIONS: This review supports surgical resection as a safe and, in the context of multimodal therapy, efficacious approach to manage HB patients who present with isolated pulmonary relapse.


Asunto(s)
Hepatoblastoma/secundario , Hepatoblastoma/cirugía , Neoplasias Hepáticas/patología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/cirugía , Metastasectomía , Neumonectomía , Preescolar , Femenino , Estudios de Seguimiento , Hepatoblastoma/mortalidad , Humanos , Lactante , Neoplasias Pulmonares/mortalidad , Masculino , Estudios Retrospectivos , Resultado del Tratamiento
9.
J Pediatr Surg ; 50(3): 382-7, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25746693

RESUMEN

BACKGROUND: Focal nodular hyperplasia (FNH) is uncommonly diagnosed in pediatric patients and may be difficult to distinguish from a malignancy. We present a review of all children with a tissue diagnosis of FNH at our institution, describe the diagnostic modalities, and provide recommendations for diagnosis and follow-up based on our experience and review of the literature. METHODS: A retrospective review of children <18years of age diagnosed with FNH at a single institution was performed from 2000 to 2013. RESULTS: Twelve patients were identified with a tissue diagnosis of FNH. Two patients required surgical resection of their lesion owing to concern for malignancy. Ten patients were managed expectantly with imaging surveillance after biopsy confirmed a diagnosis of FNH. All patients who underwent MRI had very typical findings including hypointensity on T1 weighted sequences, hyperintensity on T2, and homogenous uptake of contrast on the arterial phase. On follow-up all patients had either a stable lesion or reduction in size. CONCLUSIONS: Focal nodular hyperplasia presents typically in children with liver disease, have undergone chemotherapy, and adolescent females. Young children, particularly <5years of age, without underlying liver disease or history of chemotherapy can pose a diagnostic dilemma. In this unique subgroup of children with FNH, MRI and/or needle biopsy should be adequate diagnostic modalities for these lesions.


Asunto(s)
Diagnóstico por Imagen , Hiperplasia Nodular Focal/diagnóstico , Imagen por Resonancia Magnética/métodos , Biopsia/métodos , Niño , Diagnóstico Diferencial , Humanos
11.
Genet Med ; 17(10): 831-5, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25569436

RESUMEN

PURPOSE: We applied whole-genome sequencing (WGS) to children diagnosed with neoplasms and found to carry apparently balanced constitutional translocations to discover novel genic disruptions. METHODS: We applied the structural variation (SV) calling programs CREST, BreakDancer, SV-STAT, and CGAP-CNV, and we developed an annotative filtering strategy to achieve nucleotide resolution at the translocations. RESULTS: We identified the breakpoints for t(6;12)(p21.1;q24.31), disrupting HNF1A in a patient diagnosed with hepatic adenomas and maturity-onset diabetes of the young (MODY). Translocation as the disruptive event of HNF1A, a gene known to be involved in MODY3, has not been previously reported. In a subject with Hodgkin lymphoma and subsequent low-grade glioma, we identified t(5;18)(q35.1;q21.2), disrupting both SLIT3 and DCC, genes previously implicated in both glioma and lymphoma. CONCLUSION: These examples suggest that implementing clinical WGS in the diagnostic workup of patients with novel but apparently balanced translocations may reveal unanticipated disruption of disease-associated genes and aid in prediction of the clinical phenotype.


Asunto(s)
Estudios de Asociación Genética , Genoma Humano , Secuenciación de Nucleótidos de Alto Rendimiento , Neoplasias/genética , Translocación Genética , Adenoma/diagnóstico , Adenoma/genética , Adulto , Factores de Edad , Secuencia de Bases , Niño , Preescolar , Puntos de Rotura del Cromosoma , Cromosomas Humanos Par 12 , Cromosomas Humanos Par 18 , Cromosomas Humanos Par 5 , Cromosomas Humanos Par 6 , Receptor DCC , Variaciones en el Número de Copia de ADN , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/genética , Femenino , Genómica , Humanos , Mutación INDEL , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/genética , Masculino , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Neoplasias/diagnóstico , Polimorfismo de Nucleótido Simple , Receptores de Superficie Celular/genética , Proteínas Supresoras de Tumor/genética
12.
Pediatr Radiol ; 44(10): 1275-80, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24839140

RESUMEN

BACKGROUND: Children with hepatoblastoma routinely undergo repetitive surveillance imaging, with CT scans for several years after therapy, increasing the risk of radiation-induced cancer. OBJECTIVE: The purpose of this study was to determine the utility of surveillance CT scans compared to serum alpha-fetoprotein (AFP) levels for the detection of hepatoblastoma relapse. MATERIALS AND METHODS: This was a retrospective study of all children diagnosed with AFP-positive hepatoblastoma from 2001 to 2011 at a single institution. RESULTS: Twenty-six children with hepatoblastoma were identified, with a mean age at diagnosis of 2 years 4 months (range 3 months to 11 years). Mean AFP level at diagnosis was 132,732 ng/ml (range 172.8-572,613 ng/ml). Five of the 26 children had hepatoblastoma relapse. A total of 105 imaging exams were performed following completion of therapy; 88 (84%) CT, 8 (8%) MRI, 5 (5%) US and 4 (4%) FDG PET/CT exams. A total of 288 alpha-fetoprotein levels were drawn, with a mean of 11 per child. The AFP level was elevated in all recurrences and no relapses were detected by imaging before AFP elevation. Two false-positive AFP levels and 15 false-positive imaging exams were detected. AFP elevation was found to be significantly more specific than PET/CT and CT imaging at detecting relapse. CONCLUSION: We recommend using serial serum AFP levels as the preferred method of surveillance in children with AFP-positive hepatoblastoma, reserving imaging for the early postoperative period, for children at high risk of relapse, and for determination of the anatomical site of clinically suspected recurrence. Given the small size of this preliminary study, validation in a larger patient population is warranted.


Asunto(s)
Biomarcadores de Tumor/sangre , Hepatoblastoma/diagnóstico , Neoplasias Hepáticas/diagnóstico , Recurrencia Local de Neoplasia/diagnóstico , Tomografía Computarizada por Rayos X , alfa-Fetoproteínas/análisis , Niño , Preescolar , Femenino , Hepatoblastoma/sangre , Humanos , Lactante , Neoplasias Hepáticas/sangre , Masculino , Recurrencia Local de Neoplasia/sangre , Recurrencia , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
13.
Clin Cancer Res ; 19(23): 6578-84, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24097866

RESUMEN

PURPOSE: Imetelstat is a covalently-lipidated 13-mer thiophosphoramidate oligonucleotide that acts as a potent specific inhibitor of telomerase. It binds with high affinity to the template region of the RNA component of human telomerase (hTERC) and is a competitive inhibitor of telomerase enzymatic activity. The purpose of this study was to determine the recommended phase II dose of imetelstat in children with recurrent or refractory solid tumors. EXPERIMENTAL DESIGN: Imetelstat was administered intravenously more than two hours on days 1 and 8, every 21 days. Dose levels of 225, 285, and 360 mg/m(2) were evaluated, using the rolling-six design. Imetelstat pharmacokinetic and correlative biology studies were also performed during the first cycle. RESULTS: Twenty subjects were enrolled (median age, 14 years; range, 3-21). Seventeen were evaluable for toxicity. The most common toxicities were neutropenia, thrombocytopenia, and lymphopenia, with dose-limiting myelosuppression in 2 of 6 patients at 360 mg/m(2). Pharmacokinetics is dose dependent with a lower clearance at the highest dose level. Telomerase inhibition was observed in peripheral blood mononuclear cells at 285 and 360 mg/m(2). Two confirmed partial responses, osteosarcoma (n = 1) and Ewing sarcoma (n = 1), were observed. CONCLUSIONS: The recommended phase II dose of imetelstat given on days 1 and 8 of a 21-day cycle is 285 mg/m(2).


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Óseas/tratamiento farmacológico , Indoles/administración & dosificación , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neuroblastoma/tratamiento farmacológico , Niacinamida/análogos & derivados , Sarcoma de Ewing/tratamiento farmacológico , Adolescente , Antineoplásicos/farmacocinética , Área Bajo la Curva , Niño , Preescolar , Esquema de Medicación , Resistencia a Antineoplásicos , Femenino , Humanos , Indoles/farmacocinética , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/enzimología , Masculino , Niacinamida/administración & dosificación , Niacinamida/farmacocinética , Oligonucleótidos , Telomerasa/antagonistas & inhibidores , Telomerasa/metabolismo , Resultado del Tratamiento , Adulto Joven
14.
J Pediatr Surg ; 48(6): E33-5, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23845655

RESUMEN

Extrarenal Wilms tumors are extremely rare with only isolated case reports in the pediatric literature. We present the case of a 2-year old boy who presented with a large abdominal mass and constipation. Pathologic diagnosis of the tumor was extrarenal Wilms tumor (ERWT) with favorable histology. We discuss the diagnostic workup, radiologic and operative findings, treatment and review of the literature.


Asunto(s)
Neoplasias Retroperitoneales/diagnóstico , Tumor de Wilms/diagnóstico , Preescolar , Humanos , Región Lumbosacra , Masculino
15.
Neuro Oncol ; 15(6): 759-66, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23592571

RESUMEN

BACKGROUND: We conducted a phase I study to estimate the maximum tolerated dose and describe the dose-limiting toxicities and pharmacokinetics of oral capecitabine rapidly disintegrating tablets given concurrently with radiation therapy to children with newly diagnosed brainstem or high-grade gliomas. METHODS: Children 3-21 y with newly diagnosed intrinsic brainstem or high-grade gliomas were eligible for enrollment. The starting dose was 500 mg/m(2), given twice daily, with subsequent cohorts enrolled at 650 mg/m(2) and 850 mg/m(2) using a 3 + 3 phase I design. Children received capecitabine at the assigned dose daily for 9 wks starting from the first day of radiation therapy (RT). Following a 2-wk break, patients received 3 courses of capecitabine 1250 mg/m(2) twice daily for 14 days followed by a 7-day rest. Pharmacokinetic sampling was performed in consenting patients. Six additional patients with intrinsic brainstem gliomas were enrolled at the maximum tolerated dose to further characterize the pharmacokinetic and toxicity profiles. RESULTS: Twenty-four patients were enrolled. Twenty were fully assessable for toxicity. Dose-limiting toxicities were palmar plantar erythroderma (grades 2 and 3) and elevation of alanine aminotransferase (grades 2 and 3). Systemic exposure to capecitabine and metabolites was similar to or slightly lower than predicted based on adult data. CONCLUSIONS: Capecitabine with concurrent RT was generally well tolerated. The recommended phase II capecitabine dose when given with concurrent RT is 650 mg/m(2), administered twice daily. A phase II study to evaluate the efficacy of this regimen in children with intrinsic brainstem gliomas is in progress (PBTC-030).


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Neoplasias del Tronco Encefálico/terapia , Quimioradioterapia , Desoxicitidina/análogos & derivados , Fluorouracilo/análogos & derivados , Glioma/terapia , Comprimidos , Adolescente , Adulto , Antimetabolitos Antineoplásicos/farmacocinética , Neoplasias del Tronco Encefálico/patología , Capecitabina , Niño , Preescolar , Desoxicitidina/farmacocinética , Desoxicitidina/uso terapéutico , Femenino , Fluorouracilo/farmacocinética , Fluorouracilo/uso terapéutico , Glioma/patología , Humanos , Masculino , Dosis Máxima Tolerada , Clasificación del Tumor , Pronóstico , Dosificación Radioterapéutica , Tasa de Supervivencia , Distribución Tisular , Adulto Joven
16.
J Pediatr Hematol Oncol ; 35(4): 323-8, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23426006

RESUMEN

Peutz-Jeghers syndrome (PJS) is an autosomal dominant cancer predisposition syndrome characterized by melanotic macules and hamartomatous polyps. Small-bowel surveillance in the pediatric PJS population is not designed to identify small-bowel malignancy, which is thought to arise in adulthood. A 13-year-old boy presented with lead-point intussusception, requiring emergent surgical resection. A mucinous adenocarcinoma was found arising from high-grade dysplasia within a polyp. On the basis of these findings and mucosal pigmentation, he was diagnosed with PJS. DNA sequencing revealed a heterozygous c.921-1G>T STK11 mutation. This case is the earliest onset of small-bowel carcinoma in PJS, an observation relevant to surveillance guidelines.


Asunto(s)
Adenocarcinoma Mucinoso/etiología , Neoplasias Intestinales/etiología , Intestino Delgado/patología , Síndrome de Peutz-Jeghers/complicaciones , Adenocarcinoma Mucinoso/patología , Adenocarcinoma Mucinoso/cirugía , Adolescente , Humanos , Neoplasias Intestinales/patología , Neoplasias Intestinales/cirugía , Intestino Delgado/cirugía , Intususcepción/patología , Intususcepción/cirugía , Masculino , Síndrome de Peutz-Jeghers/patología , Síndrome de Peutz-Jeghers/cirugía
17.
Cancer Chemother Pharmacol ; 71(3): 749-63, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23314734

RESUMEN

PURPOSE: The aim of the current investigation was to develop a population pharmacokinetic model for doxorubicin and doxorubicinol that could provide improved estimated values for the pharmacokinetic parameters clearance of doxorubicin, volume of distribution of the central compartment, clearance of doxorubicinol and volume of distribution of the metabolite compartment for adults and children older than 3 years. A further aim was to investigate the potential influence of the covariates body surface area, body weight, body height, age, body mass index, sex and lean body mass on the pharmacokinetic parameters. METHODS: Three different datasets, two containing data from adults and one containing data from adults and children, were merged and the combined dataset was analysed retrospectively. In total, the combined dataset contained 934 doxorubicin and 935 doxorubicinol plasma concentrations from 82 patients [64 adults and 18 children (<18 years)]. With this combined dataset, a population pharmacokinetic model was developed, using NONMEM(®) 7.2 and a predefined model-building strategy. Different structural models, error models and estimation methods were tested, and the inter-individual and the inter-occasion variability (variability between separate (two or three) doxorubicin infusions) were tested. Using a subset of 52 patients, the influence of different covariates on the pharmacokinetic parameters was investigated. The pharmacokinetic parameter estimates obtained from doxorubicin concentrations with the best model were fixed, and an additional compartment for doxorubicinol was added to the model. With the final model for both substances, a potential age dependency and body mass index dependency of the clearance of doxorubicin and doxorubicinol as well as of the volumes of distribution of the central and the metabolite compartment were evaluated. RESULTS: A four-compartment model best described the doxorubicin and doxorubicinol data of the combined dataset. This model included a proportional residual error model and an inter-individual variability on the clearance of doxorubicin, on the inter-compartmental clearances of the peripheral compartments, on the clearance of doxorubicinol and on the volumes of distribution of the central, one peripheral and the metabolite compartment. Furthermore, the body surface area as covariate on all pharmacokinetic parameters and an inter-occasion variability for the clearance of doxorubicin and the volume of distribution of the central compartment were incorporated in the model. For a patient with the body surface area of 1.8 m², the clearance of doxorubicin was 53.3 L/h (inter-individual variability 31%, inter-occasion variability 13%) and the volume of distribution of the central compartment was 17.7 L (inter-individual variability 19%, inter-occasion variability 21%), respectively. The residual variability of the model was 22% for doxorubicin and 26% for doxorubicinol. The clearance of doxorubicinol was estimated at 44 L/h (inter-individual variability 50%) and the volume of distribution of the metabolite compartment at 1,150 L (inter-individual variability 57%). The evaluation of a possible age dependency and body mass index dependency showed a trend to a smaller volume of distribution of the central compartment (normalised to the body surface area) and a higher volume of distribution of the metabolite compartment (normalised to the body weight) in younger patients. CONCLUSIONS: A four-compartment NONMEM(®) model for doxorubicin and doxorubicinol adequately described the plasma concentrations in adults and children (>3 years). No pronounced effects of age on the clearance of doxorubicin or doxorubicinol were found, and the analysis did not support the modification of the dosing strategies presently used in children and adults.


Asunto(s)
Antibióticos Antineoplásicos/farmacocinética , Doxorrubicina/farmacocinética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/metabolismo , Análisis de Varianza , Antibióticos Antineoplásicos/administración & dosificación , Biotransformación , Índice de Masa Corporal , Niño , Preescolar , Bases de Datos Factuales , Relación Dosis-Respuesta a Droga , Doxorrubicina/administración & dosificación , Humanos , Infusiones Intravenosas , Persona de Mediana Edad , Modelos Estadísticos , Población , Reproducibilidad de los Resultados , Adulto Joven
18.
Pediatr Blood Cancer ; 60(3): 390-5, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22887890

RESUMEN

BACKGROUND: A pediatric Phase I trial was performed to determine the maximum-tolerated dose, dose-limiting toxicities (DLTs), and pharmacokinetics (PK) of vorinostat and bortezomib, in patients with solid tumors. PROCEDURE: Oral vorinostat was administered on days 1-5 and 8-12 of a 21-day cycle (starting dose 180 mg/m(2) /day with dose escalations to 230 and 300 mg/m(2) /day). Bortezomib (1.3 mg/m(2) i.v.) was administered on days 1, 4, 8, and 11 of the same cycle. PK and correlative biology studies were performed during Cycle 1. RESULTS: Twenty-three eligible patients [17 male, median age 12 years (range: 1-20)] were enrolled of whom 17 were fully evaluable for toxicity. Cycle 1 DLTs that occurred in 2/6 patients at dose level 3 (vorinostat 300 mg/m(2) /day) were Grade 2 sensory neuropathy that progressed to Grade 4 (n = 1) and Grade 3 nausea and anorexia (n = 1). No objective responses were observed. There was wide interpatient variability in vorinostat PK parameters. Bortezomib disposition was best described by a three-compartment model that demonstrated rapid distribution followed by prolonged elimination. We did not observe a decrease in nuclear factor-κB activity or Grp78 induction after bortezomib treatment in peripheral blood mononuclear cells from solid tumor patients. CONCLUSION: The recommended Phase 2 dose and schedule is vorinostat (230 mg/m(2) /day PO on days 1-5 and 8-12) in combination with bortezomib (1.3 mg/m(2) /day i.v. on days 1, 4, 8, and 11 of a 21-day cycle) in children with recurrent or refractory solid tumors.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias/tratamiento farmacológico , Adolescente , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Ácidos Borónicos/administración & dosificación , Ácidos Borónicos/efectos adversos , Ácidos Borónicos/farmacocinética , Bortezomib , Niño , Preescolar , Relación Dosis-Respuesta a Droga , Chaperón BiP del Retículo Endoplásmico , Femenino , Humanos , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/efectos adversos , Ácidos Hidroxámicos/farmacocinética , Lactante , Masculino , Dosis Máxima Tolerada , Pirazinas/administración & dosificación , Pirazinas/efectos adversos , Pirazinas/farmacocinética , Vorinostat , Adulto Joven
19.
Semin Fetal Neonatal Med ; 17(4): 216-221, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22595862

RESUMEN

Renal and hepatic tumors in neonates are extremely rare. Nevertheless it is important for clinicians to be familiar with them. Both renal and hepatic neonatal tumors are heterogeneous collections of several tumor types. Some renal and hepatic tumors are benign and may require no interventions whereas others can be associated with significant morbidity and even mortality and may require multimodality treatment. Early diagnosis and initiation of the proper treatment plan is crucial for achieving the best outcomes for these rare tumors in this vulnerable population.


Asunto(s)
Neoplasias Renales/diagnóstico , Neoplasias Hepáticas/diagnóstico , Preescolar , Diagnóstico Diferencial , Femenino , Humanos , Incidencia , Lactante , Recién Nacido , Neoplasias Renales/congénito , Neoplasias Renales/epidemiología , Neoplasias Renales/terapia , Neoplasias Hepáticas/congénito , Neoplasias Hepáticas/epidemiología , Neoplasias Hepáticas/terapia , Masculino , Pronóstico
20.
Nanomedicine (Lond) ; 7(8): 1133-48, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22583571

RESUMEN

AIM: To develop nanoshells for vascular-targeted photothermal therapy of glioma. MATERIALS & METHODS: The ability of nanoshells conjugated to VEGF and/or poly(ethylene glycol) (PEG) to thermally ablate VEGF receptor-2-positive endothelial cells upon near-infrared laser irradiation was evaluated in vitro. Subsequent in vivo studies evaluated therapy in mice bearing intracerebral glioma tumors by exposing tumors to near-infrared light after systemically delivering saline, PEG-coated nanoshells, or VEGF-coated nanoshells. The treatment effect was monitored with intravital microscopy and histology. RESULTS: VEGF-coated but not PEG-coated nanoshells bound VEGF receptor-2-positive cells in vitro to enable targeted photothermal ablation. In vivo, VEGF targeting doubled the proportion of nanoshells bound to tumor vessels and vasculature was disrupted following laser exposure. Vessels were not disrupted in mice that received saline. The normal brain was unharmed in all treatment and control mice. CONCLUSION: Nanoshell therapy can induce vascular disruption in glioma.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioma/terapia , Hipertermia Inducida/métodos , Nanocáscaras/uso terapéutico , Fototerapia/métodos , Factor A de Crecimiento Endotelial Vascular/uso terapéutico , Animales , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Células Endoteliales/metabolismo , Glioma/irrigación sanguínea , Glioma/metabolismo , Glioma/patología , Humanos , Ratones , Nanocáscaras/química , Nanocáscaras/ultraestructura , Polietilenglicoles/química , Polietilenglicoles/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA