Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pediatr Blood Cancer ; 58(2): 181-4, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21319292

RESUMEN

BACKGROUND: Circulating endothelial cells (CECs) have been detected at increased numbers in patients with solid cancers. CECs have not been systematically evaluated in patients with osteosarcoma. PROCEDURE: Patients 12 months to 30 years of age with newly diagnosed high-grade osteosarcoma were eligible for this prospective cohort study. Patients provided a single blood sample at study entry for CEC quantification by flow cytometry at a single reference laboratory. CECs were defined as CD146+, CD31+, CD45-, and CD133-. CEC progenitor cells (CEPs) were defined as CD146+, CD31+, CD45-, and CD133+. RESULTS: Eighteen patients enrolled (11 males; median age 16 years; range 5-21 years). CEC counts did not differ between patients with osteosarcoma compared to seven pediatric healthy controls (median 645 cells/ml, range 60-5,320 cells/ml vs. 1,670 cells/ml, range 330-4,700 cells/ml, respectively; P = 0.12). CEP counts did not differ between patients compared to controls (median 126 cells/ml, range 0-5,320 cells/ml vs. median 260 cells/ml, range 0-10,670 cells/ml, respectively; P = 0.69). CEC and CEP counts did not correlate with metastatic status, tumor size, or histologic response to neoadjuvant chemotherapy. CONCLUSIONS: CEC and CEP levels are not increased in patients with osteosarcoma compared to healthy controls. CECs and CEPs do not correlate with clinical features of osteosarcoma. Alternative novel markers of disease burden and response are needed in this disease.


Asunto(s)
Biomarcadores de Tumor/sangre , Neoplasias Óseas/patología , Células Endoteliales/patología , Células Neoplásicas Circulantes/patología , Osteosarcoma/patología , Células Madre/patología , Adolescente , Adulto , Antígenos CD/sangre , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Óseas/sangre , Neoplasias Óseas/tratamiento farmacológico , Estudios de Casos y Controles , Niño , Preescolar , Estudios de Cohortes , Células Endoteliales/metabolismo , Femenino , Citometría de Flujo , Estudios de Seguimiento , Humanos , Masculino , Terapia Neoadyuvante , Metástasis de la Neoplasia , Células Neoplásicas Circulantes/metabolismo , Osteosarcoma/sangre , Osteosarcoma/tratamiento farmacológico , Pronóstico , Estudios Prospectivos , Células Madre/metabolismo , Adulto Joven
2.
Cancer Res ; 66(8): 4378-84, 2006 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-16618763

RESUMEN

Much evidence supports an important role for the inducible enzyme cyclooxygenase-2 (COX-2) in tumor angiogenesis. Previous studies have focused on the role of COX-2 in stimulating endothelial proliferation, with blockade of this enzyme impairing endothelial homeostasis. However, recent data suggest that COX-2 also regulates molecules implicated in endothelial trafficking with pericytes/vascular mural cells (VMC), an interaction crucial to vessel stability. We investigated the role of COX-2 in vascular assembly by testing the effect of the specific COX-2 inhibitor SC-236 in an orthotopic xenograft model of human Wilms' tumor. Tumor growth was significantly suppressed by SC-236 (78% at day 28, 55% at day 35). Perfusion studies and immunostaining showed a marked decrease in vasculature, particularly in small vessels. Specifically, SC-236 inhibited participation of VMC in xenograft vessels. SC-236-treated tumors developed segmentally dilated, architecturally erratic tumor vessels with decreased nascent pericytes and scant mature VMC. Although vascular endothelial growth factor expression was unchanged, expression of the chemokine receptor CXCR4 was decreased in tumor vessels, consistent with defective homing of vascular progenitor cells. Vascular expression of phosphorylated platelet-derived growth factor receptor-beta was also diminished, indicating impaired VMC-endothelial trafficking. Consistent with the key role of this interaction in vessel homeostasis, vascular cells in SC-236-treated tumors displayed markedly diminished phosphorylated Akt, indicating disrupted survival signaling. These results show that SC-236 causes defective vascular assembly by attenuating incorporation of VMC into tumor vessels, impairing endothelial survival, and raise the possibility that blockade of COX-2 may provide therapeutic synergies with antiangiogenic molecules that more selectively target endothelial cells.


Asunto(s)
Inhibidores de la Ciclooxigenasa 2/farmacología , Pirazoles/farmacología , Sulfonamidas/farmacología , Tumor de Wilms/irrigación sanguínea , Tumor de Wilms/tratamiento farmacológico , Animales , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/enzimología , Neovascularización Patológica/patología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores CXCR4/biosíntesis , Receptores CXCR4/deficiencia , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Tumor de Wilms/enzimología , Tumor de Wilms/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Clin Cancer Res ; 12(17): 5190-8, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16951238

RESUMEN

PURPOSE: Angiogenesis is increased in aggressive histology non-Hodgkin's lymphoma and may be a target with selective cyclooxygenase-2 inhibition and metronomic chemotherapy. EXPERIMENTAL DESIGN: We assessed response, toxicity, and biomarkers of angiogenesis to low-dose cyclophosphamide (50 mg p.o. o.d.) and high-dose celecoxib (400 mg p.o. b.i.d.) in adult patients with relapsed or refractory aggressive non-Hodgkin's lymphoma in a multicenter phase II prospective study. RESULTS: Thirty-two of 35 patients (median age, 62 years) are evaluable for response. Patients had primarily relapsed diffuse large B-cell lymphoma (63%) were heavily pretreated (median of three regimens) and high risk (79% international prognostic index, >or=2) and 34% were relapsed after autologous stem cell transplant. With a median follow-up of 8.4 months, the overall best response rate is 37% (2 complete clinical response/complete clinical response unconfirmed and 9 partial response), with 22% achieving stable disease. Median overall and progression-free survivals are 14.4 and 4.7 months, respectively. The median response duration was 8.2 months. The most common toxicity was skin rash (40%); myelosuppression and gastrointestinal side effects were uncommon. Three patients developed deep vein thromboses and two heavily pretreated patients developed treatment-related acute myelogenous leukemia or myelodysplasia after 3.7 and 12 months of therapy. Circulating endothelial cells and their precursors declined and remained low in responders, whereas plasma vascular endothelial growth factor trended to decline in responding patients but increase in nonresponders. Trough celecoxib levels achieved targeted "antiangiogenic" levels. CONCLUSIONS: Low-dose cyclophosphamide and high-dose celecoxib is well tolerated and active in pretreated aggressive non-Hodgkin's lymphoma. Close surveillance for arterial and venous thrombotic events is recommended. The decline in circulating endothelial cells and their precursors suggests that this combination may be working by inhibiting angiogenesis but should be validated in a larger patient sample.


Asunto(s)
Ciclofosfamida/administración & dosificación , Linfoma no Hodgkin/tratamiento farmacológico , Pirazoles/administración & dosificación , Sulfonamidas/administración & dosificación , Administración Oral , Adulto , Anciano , Anciano de 80 o más Años , Celecoxib , Ciclofosfamida/efectos adversos , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Femenino , Estudios de Seguimiento , Humanos , Estimación de Kaplan-Meier , Linfoma no Hodgkin/diagnóstico , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Estudios Prospectivos , Pirazoles/efectos adversos , Pirazoles/farmacocinética , Recurrencia , Factores de Riesgo , Sulfonamidas/efectos adversos , Sulfonamidas/farmacocinética , Tasa de Supervivencia , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/sangre
4.
Clin Cancer Res ; 11(2 Pt 1): 712-9, 2005 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-15701860

RESUMEN

PURPOSE: To test the efficacy of selective therapy against cyclooxygenase-2 in combination with a low-dose regimen of a cytotoxic agent in the treatment of juvenile hematopoietic malignancies in the experimental model, Friend disease. EXPERIMENTAL DESIGN: Juvenile erythroleukemic mice (n = 8) received no treatment, celecoxib (1600 mg/kg/d), vinblastine (0.5 microg/g twice weekly), vehicle controls, or celecoxib + vinblastine combination (n = 9) over a 6-month period from time of tumor induction. Overt toxicity was assessed daily and recorded weekly. RESULTS: Among randomly selected mice from celecoxib treatment groups, plasma concentrations ranged from 2 to 6 micromol/L. As a single agent, celecoxib was not associated with any apparent toxicity. Monotherapy with vinblastine, however, caused early mortality marked by severe diarrhea, lethargy, and weight loss. At the tested doses, neither vinblastine nor celecoxib enhanced survival as monotherapies. Coadministration of these two drugs alleviated the overt toxicity associated with vinblastine and resulted in a significant increase in survival (P < 0.05). Survivors sampled throughout the study showed a trend to decreased weight loss and hematocrit levels among all groups, but significance was evidenced earlier in the vinblastine monotherapy group overall (P < 0.05). Despite similar degree of splenomegaly, histologic analysis revealed preserved splenic mantle architecture from mice given combination therapy compared with those sampled from mice on all other monotherapies, exhibiting a more diffuse burden of blasts and destruction of germinal centers. CONCLUSION: We propose that addition of a selective cyclooxygenase-2 inhibitor to a modified low-dose conventional chemotherapeutic regimen protects juvenile mice with Friend disease from succumbing to low-dose cytotoxicity, in part, by neutralizing acute inflammatory responses.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Inhibidores de la Ciclooxigenasa/uso terapéutico , Leucemia Eritroblástica Aguda/tratamiento farmacológico , Leucemia Experimental/tratamiento farmacológico , Prostaglandina-Endoperóxido Sintasas/química , Pirazoles/uso terapéutico , Sulfonamidas/uso terapéutico , Vinblastina/uso terapéutico , Animales , Enfermedades Cardiovasculares/inducido químicamente , Celecoxib , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Femenino , Virus de la Leucemia Murina de Friend/genética , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/patología , Leucemia Experimental/metabolismo , Leucemia Experimental/patología , Ratones , Ratones Endogámicos BALB C , Prostaglandina-Endoperóxido Sintasas/metabolismo , Tasa de Supervivencia , Resultado del Tratamiento
5.
Clin Pharmacol Ther ; 72(5): 490-7, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12426512

RESUMEN

BACKGROUND AND OBJECTIVE: Celecoxib is a member of a novel group of agents that selectively inhibit cyclooxygenase 2 (COX-2). COX-2 inhibitors have emerged as an important class of drugs because of the lower incidence of side effects when compared with traditional nonsteroidal anti-inflammatory drugs, which inhibit both cyclooxygenase 1 and COX-2. Because children often differ from adults with respect to drug disposition, the objective of this study was to determine the single-dose and steady-state pharmacokinetics of celecoxib in pediatric patients. METHODS: Celecoxib plasma concentrations were determined at intervals over 12 hours after a 250-mg/m(2) dose and again 1 week later after twice-daily dosing (steady state). RESULTS: Peak plasma concentrations (1234 +/- 528 microg/L) were achieved 3 hours after drug administration. The area under the celecoxib plasma concentration-time curve was 7709 +/- 3176 microg/L x h, the elimination half-life (t(1/2)) was 3.7 +/- 1.1 hours, the apparent volume of distribution was 7.9 +/- 7.8 L/kg, and the lower oral clearance of the drug was 1.4 +/- 1.0 L x h(-1) x kg(-1). Statistical analysis revealed a significantly lower [corrected] apparent oral clearance and longer t(1/2) (P <.05) at steady state compared with pharmacokinetics after a single dose. In addition, when the results were compared in children and adults, the drug was cleared approximately twice as fast in children and had a t(1/2) that was approximately half as long. CONCLUSIONS: This is the first report of celecoxib pharmacokinetics in children, and the results indicate that there are significant differences between children and adults with respect to celecoxib disposition; hence these data may have implications when dosing schedules are planned for this population.


Asunto(s)
Inhibidores de la Ciclooxigenasa/farmacocinética , Sulfonamidas/farmacocinética , Adolescente , Factores de Edad , Celecoxib , Niño , Femenino , Humanos , Masculino , Pirazoles , Sulfonamidas/administración & dosificación , Sulfonamidas/efectos adversos
6.
Pharmacotherapy ; 23(1): 97-100, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12523466

RESUMEN

Many women of reproductive age take the nonsteroidal antiinflammatory drug celecoxib. No data exist, however, regarding its transfer into human breast milk and safety for breastfed infants. We had the opportunity to obtain such data when a woman who was breastfeeding her infant daughter underwent emergency surgery to remove a gangrenous appendix. She received four doses of oral celecoxib 100 mg and did not breastfeed her daughter for 48 hours after taking the last dose. We analyzed samples of her breast milk and found a concentration of 133 ng/ml at approximately 5 hours after a 100-mg dose and an elimination half-life of 4.0-6.5 hours. This initial report, which should be substantiated and expanded, provides information that can help counsel parents about breastfeeding and celecoxib.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacocinética , Lactancia Materna , Leche Humana/metabolismo , Sulfonamidas/farmacocinética , Adulto , Antiinflamatorios no Esteroideos/administración & dosificación , Celecoxib , Femenino , Semivida , Humanos , Pirazoles , Sulfonamidas/administración & dosificación
7.
Clin Cancer Res ; 18(21): 6011-22, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22962440

RESUMEN

PURPOSE: To determine the dose-limiting toxicities (DLT), maximum tolerated dose (MTD), pharmacokinetics, and pharmacodynamics of sorafenib in children with refractory extracranial solid tumors and evaluate the tolerability of the solid tumor MTD in children with refractory leukemias. EXPERIMENTAL DESIGN: Sorafenib was administered orally every 12 hours for consecutive 28-day cycles. Pharmacokinetics (day 1 and steady-state) and pharmacodynamics were conducted during cycle 1. RESULTS: Of 65 patients enrolled, 60 were eligible. In the solid tumor cohort (n = 49), 4 of 6 patients experienced a DLT [hypertension, pain, rash/urticaria, thrombocytopenia, alanine aminotransferase (ALT)/aspartate aminotransferase (AST)] at the starting dose (150 mg/m(2)/dose) which resulted in de-escalation to 105 mg/m(2)/dose. After eligibility criteria modification and dose re-escalation, the MTD was 200 mg/m(2)/dose for solid tumors and 150 mg/m(2)/dose for leukemias. Sorafenib exposure was highly variable between patients but was within the ranges reported in adults. The apparent sorafenib clearance increased with patient age. Diarrhea, rash, fatigue, and increased ALT/AST were the most common sorafenib-related toxicities. Stable disease for 4 or more cycles was observed in 14 solid tumor patients, and 2 patients with acute myeloid leukemia (AML) and FLT3 internal tandem duplication (FLT3ITD) experienced a decrease in bone marrow blasts to less than 5%. CONCLUSIONS: The recommended phase II dose of sorafenib administered every 12 hours continuously for children with solid tumors is 200 mg/m(2)/dose and 150 mg/m(2)/dose for children with leukemias. Sorafenib toxicities and distribution in children are similar to adults. The activity of sorafenib in children with AML and FLT3ITD is currently being evaluated, and a phase II study for select solid tumors is ongoing.


Asunto(s)
Antineoplásicos/uso terapéutico , Leucemia/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Niacinamida/análogos & derivados , Compuestos de Fenilurea/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Adolescente , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Niño , Preescolar , Femenino , Humanos , Masculino , Niacinamida/farmacocinética , Niacinamida/uso terapéutico , Compuestos de Fenilurea/farmacocinética , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/farmacocinética , Sorafenib , Resultado del Tratamiento , Adulto Joven
10.
Eur J Cancer ; 46(18): 3271-9, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20656474

RESUMEN

PURPOSE: Survival rates for paediatric diffuse intrinsic brainstem glioma (DIBSG) are dismal. Metronomic dosing of temozolomide (TMZ) combined with standard radiotherapy may improve survival by increasing the therapeutic index and anti-angiogenic effect of TMZ. This study aimed to evaluate the safety and efficacy of this regimen in paediatric DIBSG patients. METHODS: Children aged 18 years or younger with newly diagnosed DIBSG were treated with standard radiotherapy and concomitant metronomic TMZ at 85 mg/m(2)/day for 6 weeks, followed by metronomic TMZ monotherapy at the same dose. Treatment was continued until tumour progression or unacceptable toxicity occurred. Primary endpoints included overall survival and toxicities. For patients who consented, plasma and urine samples were collected at diagnosis, post-induction and prior to each course of maintenance therapy for the quantification of angiogenesis markers. RESULTS: Fifteen eligible patients were enrolled, with a median age of 6.4 years. The most common toxicities were myelosuppression, most notably prolonged lymphopaenia and thrombocytopaenia. The only dose-limiting toxicity was thrombocytopaenia. Intratumoural haemorrhage was confirmed in one patient. Median time to progression was 5.13 months (95% CI = 6.4, 10.8) and median overall survival (OS) was 9.8 months (95% CI = 6.4, 10.8). Six-months OS was 80% ± 10.3%, with a 1-year OS of 20% ± 10.3%. Serum levels of both VEGF and endoglin tended to decrease during the first two cycles of therapy. CONCLUSION: Chemoradiotherapy with metronomic dosing of TMZ showed similar toxicity to previous TMZ regimens, and does not appear to improve survival in paediatric DIBSG.


Asunto(s)
Antineoplásicos Alquilantes/administración & dosificación , Neoplasias del Tronco Encefálico/tratamiento farmacológico , Neoplasias del Tronco Encefálico/radioterapia , Dacarbazina/análogos & derivados , Glioma/tratamiento farmacológico , Glioma/radioterapia , Adolescente , Antineoplásicos Alquilantes/efectos adversos , Canadá , Niño , Preescolar , Terapia Combinada/métodos , Dacarbazina/administración & dosificación , Dacarbazina/efectos adversos , Femenino , Humanos , Lactante , Masculino , Dosis Máxima Tolerada , Proyectos Piloto , Temozolomida , Trombocitopenia/etiología , Resultado del Tratamiento
11.
Neoplasia ; 11(5): 426-35, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19412427

RESUMEN

Neuroblastoma (NB) is one of the most common pediatric solid tumors originating from the neural crest lineage. Despite intensive treatment protocols including megatherapy with hematopoietic stem cell transplantation, the prognosis of NB patients remains poor. More effective therapeutics are required. High vascularity has been described as a feature of aggressive, widely disseminated NB. Our previous work demonstrated the overexpression of vascular endothelial growth factor (VEGF) in NB, and we showed that an anti-VEGF receptor (VEGFR-2) antibody could induce sustained NB tumor suppression and regression. Sunitinib is a kinase inhibitor targeting platelet-derived growth factor receptors and VEGFRs and, therefore, a promising antiangiogenic agent. In this study, we investigated the antitumor activity of sunitinib and its synergistic cytotoxicity with conventional (cyclophosphamide) and novel (rapamycin) therapies. Both NB cell lines and tumor-initiating cells from patient tumor samples were used in our in vitro and in vivo models for these drug testing. We show that sunitinib inhibits tumor cell proliferation and phosphorylation of VEGFRs. It also inhibits tumor growth, angiogenesis, and metastasis in tumor xenograft models. Low-dose sunitinib (20 mg/kg) demonstrates synergistic cytotoxicity with an mTOR inhibitor, rapamycin, which is more effective than the traditional chemotherapeutic drug, cyclophosphamide. These preclinical studies provide the evidence of antitumor activity of sunitinib both in the early stage of tumor formation and in the progressive metastatic disease. These studies also provide the framework for clinical trial of sunitinib, alone and in combination with conventional and novel therapies to increase efficacy and improve patient outcome in NB.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Indoles/farmacología , Neovascularización Patológica/tratamiento farmacológico , Neuroblastoma/tratamiento farmacológico , Pirroles/farmacología , Animales , Western Blotting , Línea Celular Tumoral , Modelos Animales de Enfermedad , Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Ratones , Neovascularización Patológica/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Sunitinib , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Eur J Cancer ; 45(13): 2352-9, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19505817

RESUMEN

PURPOSE: To evaluate the safety, efficacy and pharmacokinetics of imatinib in children with recurrent or refractory central nervous system (CNS) tumours expressing KIT and/or PDGFRA. METHODS: Nineteen patients aged 2-18 years, with recurrent or refractory CNS tumours expressing either of the target receptors KIT and/or PDGFRA (by immunohistochemistry) were eligible. Participants received imatinib orally at a dose of 440 mg/m(2)/day and toxicities and tumour responses were monitored. Serial blood and cerebrospinal fluid samples for pharmacokinetics were obtained in a subset of consenting patients. Frozen tumour samples were analysed retrospectively for KIT and PDGFRA gene amplification in a subset of patients for whom samples were available. RESULTS: Common toxicities were lymphopaenia, neutropaenia, leucopaenia, elevated serum transaminases and vomiting. No intratumoural haemorrhages were observed. Although there were no objective responses to imatinib, four patients had long-term stable disease (SD) (38-104 weeks). Our results suggest a possible relationship between KIT expression and maintenance of SD with imatinib treatment; KIT immunopositivity was seen in only 58% (11/19) of study participants overall, but in 100% of patients with SD at 38 weeks. All patient tumours showed PDGFRA expression. Pharmacokinetic data showed a high interpatient variability, but corresponded with previously reported values. CONCLUSIONS: Imatinib at 440 mg/m(2)/day is relatively safe in children with recurrent CNS tumours, but induced no objective responses. Demonstration of SD in previously progressing patients (KIT-expressing) suggests cytostatic activity of imatinib.


Asunto(s)
Antineoplásicos/farmacocinética , Neoplasias del Sistema Nervioso Central/metabolismo , Piperazinas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacocinética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Pirimidinas/farmacocinética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Adolescente , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Benzamidas , Canadá , Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Neoplasias del Sistema Nervioso Central/genética , Niño , Preescolar , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Mesilato de Imatinib , Masculino , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Piperazinas/administración & dosificación , Piperazinas/efectos adversos , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/efectos adversos , Proteínas Proto-Oncogénicas c-kit/genética , Pirimidinas/administración & dosificación , Pirimidinas/efectos adversos , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética
13.
J Clin Oncol ; 26(3): 399-405, 2008 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-18202416

RESUMEN

PURPOSE: We conducted a pediatric phase I trial of the vascular endothelial growth factor (VEGF)-neutralizing antibody bevacizumab (BV). Primary aims included estimating the maximum-tolerated dose (MTD) and determining the dose-limiting toxicities (DLTs), pharmacokinetics, and biologic effects of BV in children with cancer. PATIENTS AND METHODS: BV (5, 10, 15 mg/kg) was administered intravenously every 2 weeks in 28-day courses to children with refractory solid tumors. RESULTS: Twenty-one patients enrolled, 20 (median age, 13 years) were eligible, and 18 completed one course and were fully assessable for toxicity. A total of 67 courses were administered (median, three courses per patient; range, one to 16 courses). Treatment was well tolerated with no DLTs observed. Non-DLTs included infusional reaction, rash, mucositis, proteinuria, and lymphopenia. Increases in systolic and diastolic blood pressure not meeting Common Terminology Criteria for Adverse Events (CTCAEv3) pediatric-specific criteria for hypertension were observed. There was no hemorrhage or thrombosis. Growth perturbation was not detected in a limited sample over the first course. The serum exposure to BV as measured by area under the concentration-time curve (AUC) seemed to increase in proportion to dose. The median clearance of BV was 4.1 mL/d/kg (range, 3.1 to 15.5 mL/d/kg), and the median half-life was 11.8 days (range, 4.4 to 14.6 days). No objective responses were observed. Exploratory analyses on circulating endothelial mobilization and viability are consistent with the available adult data. CONCLUSION: BV is well tolerated in children. Phase II pediatric studies of BV in combination with chemotherapy in dosing schedules similar to adults are planned.


Asunto(s)
Inhibidores de la Angiogénesis/farmacocinética , Anticuerpos Monoclonales/farmacocinética , Neoplasias/metabolismo , Adolescente , Adulto , Inhibidores de la Angiogénesis/efectos adversos , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales Humanizados , Área Bajo la Curva , Bevacizumab , Niño , Preescolar , Femenino , Semivida , Humanos , Lactante , Infusiones Intravenosas , Masculino , Dosis Máxima Tolerada , Estadificación de Neoplasias , Neoplasias/patología , Factores de Riesgo , Tasa de Supervivencia , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
14.
J Pediatr Hematol Oncol ; 29(1): 32-47, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17230065

RESUMEN

The high prevalence of complementary and alternative medicine use including natural health products (NHPs) in the pediatric oncology population is well established. The potential for concurrent use of NHPs with conventional chemotherapy necessitates physician awareness regarding the potential risks and benefits that might come from this coadministration. Knowledge of interactions between NHPs and chemotherapy is poorly characterized; however, an understanding of potential mechanisms of interaction by researchers and clinicians is important. Concerns regarding the use of antioxidants during chemotherapy are controversial and evidence exists to support both adherents and detractors in this debate. Our review addresses issues regarding potential interactions between NHPs and chemotherapies used in pediatric oncology from a pharmacokinetic and pharmacodynamic perspective. Examples of combinations of NHP and chemotherapies are briefly presented in addition to a strategy to avoid (or induce) a possible interaction between a NHP and chemotherapy. In conclusion, more clinical research is needed to substantiate or preclude the use of NHPs in the treatment of cancer and especially in combination with chemotherapy.


Asunto(s)
Productos Biológicos , Terapias Complementarias , Interacciones Farmacológicas , Neoplasias/tratamiento farmacológico , Adolescente , Productos Biológicos/agonistas , Productos Biológicos/antagonistas & inhibidores , Productos Biológicos/farmacocinética , Productos Biológicos/uso terapéutico , Niño , Preescolar , Ensayos Clínicos como Asunto , Terapias Complementarias/efectos adversos , Terapias Complementarias/métodos , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Neoplasias/complicaciones
15.
Pediatrics ; 119(6): e1239-47, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17485449

RESUMEN

OBJECTIVES: Oral systemic corticosteroids are the mainstay of treatment for problematic hemangiomas; however, current information is based on anecdotal experience and retrospective studies. We aimed to determine whether systemic steroids are efficacious in proliferating hemangioma and to compare the efficacy and safety of 2 corticosteroid treatment modalities. PATIENTS AND METHODS: Twenty patients with problematic hemangiomas of infancy were randomly assigned to either daily oral prednisolone or monthly intravenous pulses of methylprednisolone. Their clinical outcomes (improvement using a visual analog score) and adverse events were compared at 3 months from baseline and 1 year of age. Data on possible surrogate markers of angiogenesis were available for the first 3 months. RESULTS: At 3 months, orally treated patients had a median visual analog score of 70 compared with 12 in the intravenous group. This response pattern was similar at the patients' first birthday: 50.0 vs -1.5. Additional treatment beyond 3 months was needed for 65% of the patients (7 in the intravenous and 6 in the oral group). Six of 8 patients with impaired vision at enrollment had an improved function at 1 year (4 patients in the intravenous group and 3 patients in the oral group). Of the 4 surrogate markers of angiogenesis measured (plasma basic fibroblast growth factor, vascular endothelial growth factor, vascular cellular adhesion molecule 1, endoglin, and urine basic fibroblast growth factor), the only 2 that decreased over time were vascular cellular adhesion molecule 1 and endoglin. Patients in the oral group had a higher rate of adverse effects, such as hypertension (18.6% vs 13.1%), abnormal cortisol (78% vs 60%), and growth retardation. CONCLUSIONS: Systemic corticosteroids are efficacious in stopping the proliferation of hemangiomas. The oral corticosteroids offered more clinical and biological benefit than the pulse steroids with higher risk of adverse effects.


Asunto(s)
Corticoesteroides/administración & dosificación , Hemangioma/tratamiento farmacológico , Administración Oral , Corticoesteroides/efectos adversos , Biomarcadores/sangre , Femenino , Hemangioma/sangre , Hemangioma/patología , Humanos , Hipertensión/inducido químicamente , Lactante , Inyecciones Intravenosas , Masculino , Dimensión del Dolor/efectos de los fármacos , Síndrome de Dificultad Respiratoria/inducido químicamente , Molécula 1 de Adhesión Celular Vascular/sangre
16.
Cancer Invest ; 24(4): 432-43, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16777697

RESUMEN

Pediatric cancer has a better outcome profile than adult cancers. However, refractory disease and the potential for long-term morbidity resulting from the use of conventional therapies necessitate the development of novel treatments for this population. Recent advances in oncology include the use of low dose metronomic (LDM) chemotherapy. The promise of this novel therapeutic approach includes reduced toxicity and the potential for efficacy predominantly through an antiangiogenic effect. The clinical benefit may be realized especially when combined with other antiangiogenic agents and/or conventional maximally tolerated doses of chemotherapy. In this article, we review the evidence for the use of LDM chemotherapy with a focus on pediatric cancer. Included are some of the possible risks attributable to this therapy in a pediatric setting and some of the hurdles to overcome in order to conduct good clinical research. Emphasis is placed on the development of proper surrogate markers to monitor antiangiogenic therapy in order to both optimize the dosing schedule for LDM chemotherapy and to provide a way of tracking therapeutic efficacy.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Antineoplásicos/administración & dosificación , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Adulto , Animales , Biomarcadores de Tumor/análisis , Niño , Esquema de Medicación , Humanos , Dosis Máxima Tolerada
17.
J Pediatr Hematol Oncol ; 28(11): 720-8, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17114958

RESUMEN

Tumor vasculature is a reasonable target for cancer therapy and lower more frequent doses of traditional chemotherapeutics [low-dose metronomic (LDM) chemotherapy] has been shown to have antiangiogenic efficacy. This study evaluated the safety and pharmacokinetics of celecoxib and LDM vinblastine or cyclophosphamide in children with recurrent, refractory solid tumors. We also investigated whether a subset of circulating plasma proteins are surrogate markers of angiogenic activity. Thirty-three children were enrolled in this pilot study and received celecoxib (250 mg/m(2) PO b.i.d.) and either vinblastine (1 mg/m(2) IV 3 x /wk) or cyclophosphamide (30 mg/m(2) PO daily) continually. Celecoxib alone and with LDM chemotherapy was well tolerated and plasma concentrations were consistent with those shown to have antiangiogenic activity. Four patients (13%) had durable stable disease (28 to 78 wk) although no complete or partial responses were observed. The surrogate markers measured (vascular endothelial growth factor, basic fibroblast growth factor, soluble vascular cell adhesion molecule, soluble intercellular cell adhesion molecule, endostatin, and thrombospondin-1) were highly variable and no statistically significant relationship between them and disease progression or maintenance of stable disease was observed. We concluded that this regimen is well tolerated hence supporting the use of this form of therapy in pediatric patients. However, future studies should include more homogenous patient populations and focus on validating surrogate markers to monitor treatment activity.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ciclofosfamida/administración & dosificación , Neoplasias/tratamiento farmacológico , Pirazoles/administración & dosificación , Sulfonamidas/administración & dosificación , Vinblastina/administración & dosificación , Adolescente , Inhibidores de la Angiogénesis , Biomarcadores/análisis , Celecoxib , Niño , Preescolar , Esquema de Medicación , Femenino , Humanos , Masculino , Recurrencia Local de Neoplasia , Neoplasias/metabolismo , Proyectos Piloto , Pirazoles/efectos adversos , Sulfonamidas/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA