Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Pediatr Blood Cancer ; 70(7): e30365, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37073741

RESUMO

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.


Assuntos
Hepatoblastoma , Neoplasias Hepáticas , Criança , Humanos , Hepatoblastoma/patologia , Irinotecano/uso terapêutico , Vincristina , Neoplasias Hepáticas/patologia , alfa-Fetoproteínas , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Resultado do Tratamento
2.
Cancer ; 128(5): 1057-1065, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-34762296

RESUMO

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.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica , Hepatoblastoma , Neoplasias Hepáticas , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Cisplatino/efeitos adversos , Doxorrubicina/efeitos adversos , Estudos de Viabilidade , Hepatoblastoma/tratamento farmacológico , Hepatoblastoma/patologia , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Resultado do Tratamento , Vincristina/efeitos adversos
3.
J Pediatr Hematol Oncol ; 39(1): 72-76, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27820134

RESUMO

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.


Assuntos
Algoritmos , Metotrexato/administração & dosagem , Sistemas Automatizados de Assistência Junto ao Leito , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/prevenção & controle , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Área Sob a Curva , Relação Dose-Resposta a Droga , Esquema de Medicação , Monitoramento de Medicamentos , Hidratação , Humanos , Infusões Intravenosas , Metotrexato/efeitos adversos , Metotrexato/farmacocinética , Mucosite/induzido quimicamente , Mucosite/prevenção & controle , Doenças do Sistema Nervoso/induzido quimicamente , Doenças do Sistema Nervoso/prevenção & controle , Estudos Retrospectivos
4.
J Hepatol ; 65(2): 325-33, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27117591

RESUMO

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.


Assuntos
Neoplasias Hepáticas , Animais , Carcinoma Hepatocelular , Linhagem Celular Tumoral , Criança , Modelos Animais de Doenças , Xenoenxertos , Humanos , Camundongos , Transplante de Neoplasias , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Genet Med ; 17(10): 831-5, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25569436

RESUMO

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.


Assuntos
Estudos de Associação Genética , Genoma Humano , Sequenciamento de Nucleotídeos em Larga Escala , Neoplasias/genética , Translocação Genética , Adenoma/diagnóstico , Adenoma/genética , Adulto , Fatores Etários , Sequência de Bases , Criança , Pré-Escolar , Pontos de Quebra do Cromossomo , Cromossomos Humanos Par 12 , Cromossomos Humanos Par 18 , Cromossomos Humanos Par 5 , Cromossomos Humanos Par 6 , Receptor DCC , Variações do Número de Cópias de DNA , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/genética , Feminino , Genômica , Humanos , Mutação INDEL , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/genética , Masculino , Proteínas de Membrana/genética , Dados de Sequência Molecular , Neoplasias/diagnóstico , Polimorfismo de Nucleotídeo Único , Receptores de Superfície Celular/genética , Proteínas Supressoras de Tumor/genética
6.
Pediatr Radiol ; 44(10): 1275-80, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24839140

RESUMO

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.


Assuntos
Biomarcadores Tumorais/sangue , Hepatoblastoma/diagnóstico , Neoplasias Hepáticas/diagnóstico , Recidiva Local de Neoplasia/diagnóstico , Tomografia Computadorizada por Raios X , alfa-Fetoproteínas/análise , Criança , Pré-Escolar , Feminino , Hepatoblastoma/sangue , Humanos , Lactente , Neoplasias Hepáticas/sangue , Masculino , Recidiva Local de Neoplasia/sangue , Recidiva , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
7.
Pediatr Blood Cancer ; 60(3): 390-5, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22887890

RESUMO

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.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias/tratamento farmacológico , Adolescente , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Ácidos Borônicos/administração & dosagem , Ácidos Borônicos/efeitos adversos , Ácidos Borônicos/farmacocinética , Bortezomib , Criança , Pré-Escolar , Relação Dose-Resposta a Droga , Chaperona BiP do Retículo Endoplasmático , Feminino , Humanos , Ácidos Hidroxâmicos/administração & dosagem , Ácidos Hidroxâmicos/efeitos adversos , Ácidos Hidroxâmicos/farmacocinética , Lactente , Masculino , Dose Máxima Tolerável , Pirazinas/administração & dosagem , Pirazinas/efeitos adversos , Pirazinas/farmacocinética , Vorinostat , Adulto Jovem
8.
J Pediatr Hematol Oncol ; 35(4): 323-8, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23426006

RESUMO

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.


Assuntos
Adenocarcinoma Mucinoso/etiologia , Neoplasias Intestinais/etiologia , Intestino Delgado/patologia , Síndrome de Peutz-Jeghers/complicações , Adenocarcinoma Mucinoso/patologia , Adenocarcinoma Mucinoso/cirurgia , Adolescente , Humanos , Neoplasias Intestinais/patologia , Neoplasias Intestinais/cirurgia , Intestino Delgado/cirurgia , Intussuscepção/patologia , Intussuscepção/cirurgia , Masculino , Síndrome de Peutz-Jeghers/patologia , Síndrome de Peutz-Jeghers/cirurgia
9.
Curr Oncol Rep ; 14(2): 197-205, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22302601

RESUMO

Pediatric soft-tissue and bone sarcomas are a heterogeneous group of tumors of mesenchymal origin which affect approximately 1,500 children in the United States each year. Using multimodal therapy (surgery, radiation, and chemotherapy),the overall 5-year survival rate for children with soft-tissue and bone sarcomas is approximately 60%­70%. However, the prognosis for children with metastatic or recurrent disease is poor; and, furthermore, the improvements in the overall cure rate have slowed. It is highly unlikely that further advances in the treatment of pediatric soft-tissue and bone sarcomas will come from traditional cytotoxic chemotherapy. Based on research advances in understanding the biology of pediatric soft-tissue and bone sarcomas, improved cure rates will likely be driven by new types of treatment which target the specific abnormalities within these tumors. These new targeted therapies may include small molecules, antibodies, or other immunotherapies. This review briefly describes the biology of the major types of pediatric sarcomas, discusses potential targets for new therapy, and highlights some recent and current clinical trials using targeted therapy.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Ósseas/tratamento farmacológico , Terapia de Alvo Molecular/métodos , Sarcoma/tratamento farmacológico , Neoplasias de Tecidos Moles/tratamento farmacológico , Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Adolescente , Inibidores da Angiogênese/uso terapêutico , Criança , Pré-Escolar , Ensaios Clínicos como Assunto , Humanos , Prognóstico , Inibidores de Proteínas Quinases/uso terapêutico
10.
J Neurooncol ; 104(1): 55-63, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21110217

RESUMO

We are developing a novel treatment for high-grade gliomas using near infrared-absorbing silica-gold nanoshells that are thermally activated upon exposure to a near infrared laser, thereby irreversibly damaging cancerous cells. The goal of this work was to determine the efficacy of nanoshell-mediated photothermal therapy in vivo in murine xenograft models. Tumors were induced in male IcrTac:ICR-Prkdc(SCID) mice by subcutaneous implantation of Firefly Luciferase-labeled U373 human glioma cells and biodistribution and survival studies were performed. To evaluate nanoparticle biodistribution, nanoshells were delivered intravenously to tumor-bearing mice and after 6, 24, or 48 h the tumor, liver, spleen, brain, muscle, and blood were assessed for gold content by inductively coupled plasma-mass spectrometry (ICP-MS) and histology. Nanoshell concentrations in the tumor increased for the first 24 h and stabilized thereafter. Treatment efficacy was evaluated by delivering saline or nanoshells intravenously and externally irradiating tumors with a near infrared laser 24 h post-injection. Success of treatment was assessed by monitoring tumor size, tumor luminescence, and survival time of the mice following laser irradiation. There was a significant improvement in survival for the nanoshell treatment group versus the control (P < 0.02) and 57% of the mice in the nanoshell treatment group remained tumor free at the end of the 90-day study period. By comparison, none of the mice in the control group survived beyond 24 days and mean survival was only 13.3 days. The results of these studies suggest that nanoshell-mediated photothermal therapy represents a promising novel treatment strategy for malignant glioma.


Assuntos
Neoplasias Encefálicas/terapia , Glioma/terapia , Nanoconchas/uso terapêutico , Fototerapia/métodos , Animais , Neoplasias Encefálicas/mortalidade , Linhagem Celular Tumoral , Modelos Animais de Doenças , Glioma/mortalidade , Humanos , Estimativa de Kaplan-Meier , Medições Luminescentes , Camundongos , Transplante de Neoplasias/métodos , Espectrometria de Massas em Tandem/métodos , Fatores de Tempo , Distribuição Tecidual
12.
Pediatr Blood Cancer ; 52(5): 621-5, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19137570

RESUMO

BACKGROUND: Hemophagocytic Lymphohistiocytosis (HLH) is characterized by uncontrolled inflammation that is generally fatal without immune modulating chemotherapy. At Texas Children's Hospital, we have observed significant central nervous system (CNS) toxicity in several patients treated for HLH according to the Histiocyte Society protocol HLH-2004 in which cyclosporine is given early in the treatment regimen. METHODS: Patients diagnosed with HLH at Texas Children's Hospital between April 2004 and October 2007 were identified and charts were reviewed. A reference group of patients treated between August 2001 and March 2004, prior to the introduction of HLH-2004, was also evaluated. RESULTS: Five of 17 patients in the study group developed severe neurotoxicity. Four had new onset seizures associated with significant MRI abnormalities, while the fifth died of intracerebral hemorrhage. Timing of the development of neurologic side effects ranged from day 5 to week 6 of therapy. Cyclosporine levels were outside the therapeutic range (200-300 ng/ml) prior to the onset of symptoms in two of the five patients. Systolic blood pressures for all five patients were greater than the 95th percentile for age on at least one measurement within 24 hr of the onset of neurologic symptoms. MRI scans obtained within 24 hr of seizure activity in four patients were consistent with posterior reversible encephalopathy syndrome (PRES). By comparison only one patient in the reference group (n = 15) had neurotoxicity (PRES). CONCLUSIONS: Patients being treated for HLH appear to be at risk for neurotoxicity, particularly PRES. Elevated blood pressure, worsening renal and liver function, increased cyclosporine levels, and CNS involvement of HLH may be triggers for the neurotoxic side effects of treatment. Patients being treated on HLH-2004 require close monitoring of their neurologic status and modifiable risk factors such as hypertension should managed aggressively. If larger studies validate our observations, it will be important to determine if up-front cyclosporine in HLH protocols confers a survival benefit that outweighs the potential risk of increased neurotoxicity.


Assuntos
Doenças do Sistema Nervoso Central/induzido quimicamente , Doenças do Sistema Nervoso Central/patologia , Ciclosporina/efeitos adversos , Ciclosporina/uso terapêutico , Linfo-Histiocitose Hemofagocítica/tratamento farmacológico , Adolescente , Doença Hepática Induzida por Substâncias e Drogas , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Hepatopatias/fisiopatologia , Imageamento por Ressonância Magnética , Masculino , Modelos Biológicos
13.
Clin Pharmacokinet ; 58(7): 899-910, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30810947

RESUMO

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.


Assuntos
Antimetabólitos Antineoplásicos/farmacocinética , Metotrexato/farmacocinética , Modelos Biológicos , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Feminino , Humanos , Lactente , Masculino , Medicina de Precisão
14.
Cancer Chemother Pharmacol ; 83(2): 349-360, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30488179

RESUMO

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.


Assuntos
Algoritmos , Antimetabólitos Antineoplásicos/administração & dosagem , Antimetabólitos Antineoplásicos/farmacocinética , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Metotrexato/administração & dosagem , Metotrexato/farmacocinética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Adolescente , Adulto , Criança , Pré-Escolar , Relação Dose-Resposta a Droga , Feminino , Seguimentos , Humanos , Lactente , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Prognóstico , Estudos Prospectivos , Fatores de Risco , Distribuição Tecidual , Adulto Jovem
15.
Cancer Chemother Pharmacol ; 61(4): 647-52, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17541590

RESUMO

PURPOSE: Valproic acid (VPA), a widely used antiepileptic, also inhibits histone deacetylase (HDAC), and is undergoing evaluation as an anti-cancer agent. We studied the pharmacokinetics of VPA in the plasma and cerebrospinal fluid (CSF) in a non-human primate model that is highly predictive of human CSF penetration to determine if levels of VPA required to inhibit HDAC in in vivo models can be attained. METHODS: Oral VPA, 75 mg/kg, was administered to four non-human primates. Serial samples of blood (n = 4) and CSF (n = 3) were obtained for pharmacokinetic studies of total and free VPA. Plasma and CSF VPA concentrations were measured using the commercially available Abbott AxSYM VPA assay reagent system (Abbott Laboratories, Abbott Park, IL, USA). The resultant plasma and CSF data were evaluated using pharmacokinetic modeling methods. RESULTS: At a dose of 75 mg/kg, the maximum plasma concentration of VPA was 130.1 +/- 70.6 microg/ml (mean +/- standard deviation) for total drug and 53.3 +/- 44.4 microg/ml for free drug. The mean plasma area under the curve (AUC) for total drug was 680 +/- 233 microg/ml h and for free drug 146 +/- 89 microg/ml hr. The maximum CSF concentration occurred 2-3 h after administration and was 28.2 +/- 18.6 microg/ml. The CSF AUC for VPA was 108 +/- 52 microg/ml h. The CSF penetration of VPA was 12.9 +/- 5.1% for total drug and 57.0 +/- 8.7% for free drug. Disappearance from the plasma followed non-linear kinetics with a V (max) of 321.2 +/- 65.6 microg/kg/min and a K (m) of 17.2 +/- 13.7 mg/l. CONCLUSION: Valproic acid deserves further study for the treatment of CNS tumors given its high CSF penetration after oral dosing coupled with the anti-tumor activity observed in preclinical studies.


Assuntos
Antineoplásicos/farmacocinética , Ácido Valproico/farmacocinética , Animais , Antineoplásicos/sangue , Antineoplásicos/líquido cefalorraquidiano , Área Sob a Curva , Inibidores Enzimáticos/sangue , Inibidores Enzimáticos/líquido cefalorraquidiano , Inibidores Enzimáticos/farmacocinética , Meia-Vida , Inibidores de Histona Desacetilases , Macaca mulatta , Ácido Valproico/sangue , Ácido Valproico/líquido cefalorraquidiano
16.
Clin Cancer Res ; 13(5): 1516-22, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17332297

RESUMO

PURPOSE: A phase 1 study to determine the maximum-tolerated dose, dose-limiting toxicity, pharmacokinetics, and biological effects of bortezomib in children with recurrent/refractory leukemia. EXPERIMENTAL DESIGN: Bortezomib was administered twice weekly for 2 consecutive weeks at either 1.3 or 1.7 mg/m(2) dose followed by a 1-week rest. Bortezomib pharmacokinetics and nuclear factor kappaB (NF-kappaB) binding activity were evaluated during the first treatment cycle. RESULTS: Twelve patients (nine with acute lymphoblastic leukemia, three with acute myelogenous leukemia), median age 11 years (range, 1-18 years), were enrolled between May 2004 and November 2005, of whom seven were not fully evaluable for toxicity due to rapidly progressive disease or uncontrolled infection. Dose-limiting toxicities occurred in two patients at the 1.7 mg/m(2) dose level. One patient experienced grade 3 confusion and the other patient had grade 4 febrile neutropenia associated with grade 4 hypotension and grade 3 creatinine. Pharmacokinetic analysis at 1.3 mg/m(2) revealed a clearance of 11 mL/h/m(2), a central volume of distribution of 6.7 L/m(2), and a terminal half-life of 12.6 h. NF-kappaB activity was examined in five patients and was noted to transiently increase and then decrease 4- to 6-fold by 24 h following bortezomib in two patients. There were no objective clinical responses. CONCLUSIONS: For children with leukemia, the recommended phase 2 dose of bortezomib, administered twice weekly for 2 weeks followed by a 1-week rest, is 1.3 mg/m(2)/dose. Although bortezomib treatment inhibited NF-kappaB activity, bortezomib had little activity as a single agent in this population.


Assuntos
Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Ácidos Borônicos/farmacocinética , Ácidos Borônicos/uso terapêutico , Leucemia/tratamento farmacológico , Pirazinas/farmacocinética , Pirazinas/uso terapêutico , Adolescente , Bortezomib , Criança , Pré-Escolar , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Quinase I-kappa B/efeitos dos fármacos , Immunoblotting , Lactente , Masculino , Dose Máxima Tolerável , NF-kappa B/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos
17.
Cancer Chemother Pharmacol ; 59(4): 461-6, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16855840

RESUMO

PURPOSE: Pemetrexed, a multi-targeted antifolate that disrupts synthesis of both purines and pyrimidines, is approved for use in malignant pleural mesothelioma and non-small cell lung cancer. Pemetrexed is currently being evaluated for anti-tumor activity in a variety of solid and central nervous system tumors. We studied the plasma and cerebrospinal fluid (CSF) pharmacokinetics of pemetrexed in a non-human primate model that is highly predictive of human CSF penetration. METHODS: Pemetrexed, 20 mg/kg (400 mg/m2), was administered intravenously to four non-human primates. Serial blood samples were obtained from all animals and serial CSF samples were obtained from three animals. Plasma and CSF concentrations of pemetrexed were measured using LC/MS/MS and the resulting concentration versus time data were evaluated using model independent and dependent methods. RESULTS: Pemetrexed disappearance from plasma was best described by a two compartment model with a mean distribution half-life of 13.8 +/- 3.2 min and an elimination half-life of 70.0 +/- 16.0 min. The volume of distribution of and the clearance from the central compartment were 0.066 +/- 0.017 l/kg and 3.6 +/- 0.6 ml/min/kg, respectively. Peak CSF concentrations occurred 40-71 min after the start of the infusion with an average of 0.26 +/- 0.15 microM. CONCLUSION: The CSF penetration of pemetrexed was less than 2% (range 0.33-1.58%), suggesting that it should be used in conjunction with other CNS preventive strategies when used in the treatment of malignancies with a predilection for CNS or leptomeningeal metastases.


Assuntos
Antimetabólitos Antineoplásicos/farmacocinética , Antagonistas do Ácido Fólico/farmacocinética , Glutamatos/farmacocinética , Guanina/análogos & derivados , Animais , Área Sob a Curva , Glutamatos/administração & dosagem , Guanina/administração & dosagem , Guanina/farmacocinética , Meia-Vida , Infusões Intravenosas , Macaca mulatta , Pemetrexede , Ligação Proteica
18.
Cancer Chemother Pharmacol ; 59(6): 847-53, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17136402

RESUMO

PURPOSE: We performed a pharmacokinetic evaluation of methotrexate (MTX) in infants with acute lymphoblastic leukemia enrolled on the Pediatric Oncology Group (POG) 9407 Infant Leukemia Study to evaluate the effects of age on MTX pharmacokinetics and pharmacodynamics. METHODS: A pharmacokinetic database of 61 patients was developed by combining MTX data obtained from 16 patients in a pharmacokinetic sub-study with data obtained for clinical care in other patients enrolled on the POG 9407 protocol. The data were analyzed for the first dose of MTX given to patients in induction/intensification therapy. Patients received MTX (4 g/m2) over 24 h at week 4 of therapy. Toxicity data were also reviewed to evaluate the incidence of common MTX toxicities during the first 6 weeks of therapy (the induction/intensification phase). RESULTS: Steady-state clearance (mean+/-standard deviation) for infants aged 0-6 months was 89+/-32 ml/min/m2 compared to 111+/-40 for infants aged 7-12 months (P=0.030). In the subgroup of infants aged 0-3 months the mean steady-state clearance was 84+/-30 ml/min/m2 (P=0.026 vs. the 7-12-month group). The incidence of renal toxicity (all grades) during induction/intensification therapy was 23% in the 0-3 months age group compared to 0% (for n=27) in the group 7-12 months of age (P=0.029). There were no significant differences in hepatoxicity or mucous membrane toxicity between age groups. CONCLUSIONS: A modest difference in steady-state MTX clearance is observed between younger infants (0-6 months) and older infants (7-12 months). Very young infants (0-3 months) also experienced a slightly higher incidence of renal toxicity during induction/intensification therapy. Steady-state clearance for the older infants is similar to values reported for children in other studies.


Assuntos
Metotrexato/farmacocinética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Fatores Etários , Esquema de Medicação , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Metotrexato/efeitos adversos , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo
20.
Cancer Chemother Pharmacol ; 79(1): 181-187, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28032129

RESUMO

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.


Assuntos
Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacocinética , Pirazóis/farmacocinética , Piridinas/farmacocinética , Administração Oral , Adolescente , Adulto , Criança , Pré-Escolar , Crizotinibe , Feminino , Humanos , Linfoma Anaplásico de Células Grandes/tratamento farmacológico , Masculino , Inibidores de Proteínas Quinases/administração & dosagem , Pirazóis/administração & dosagem , Piridinas/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA