Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Clin Oncol ; 42(1): 90-102, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-37883734

RESUMEN

PURPOSE: Long-term survival in high-risk neuroblastoma (HRNB) is approximately 50%, with mortality primarily driven by relapse. Eflornithine (DFMO) to reduce risk of relapse after completion of immunotherapy was investigated previously in a single-arm, phase II study (NMTRC003B; ClinicalTrials.gov identifier: NCT02395666) that suggested improved event-free survival (EFS) and overall survival (OS) compared with historical rates in a phase III trial (Children Oncology Group ANBL0032; ClinicalTrials.gov identifier: NCT00026312). Using patient-level data from ANBL0032 as an external control, we present new analyses to further evaluate DFMO as HRNB postimmunotherapy maintenance. PATIENTS AND METHODS: NMTRC003B (2012-2016) enrolled patients with HRNB (N = 141) after standard up-front or refractory/relapse treatment who received up to 2 years of continuous treatment with oral DFMO (750 ± 250 mg/m2 twice a day). ANBL0032 (2001-2015) enrolled patients with HRNB postconsolidation, 1,328 of whom were assigned to dinutuximab (ch.14.18) treatment. Selection rules identified 92 NMTRC003B patients who participated in (n = 87) or received up-front treatment consistent with (n = 5) ANBL0032 (the DFMO/treated group) and 852 patients from ANBL0032 who could have been eligible for NMTRC003B after immunotherapy, but did not enroll (the NO-DFMO/control group). The median follow-up time for DFMO/treated patients was 6.1 years (IQR, 5.2-7.2) versus 5.0 years (IQR, 3.5-7.0) for NO-DFMO/control patients. Kaplan-Meier and Cox regression compared EFS and OS for overall groups, 3:1 (NO-DFMO:DFMO) propensity score-matched cohorts balanced on 11 baseline demographic and disease characteristics with exact matching on MYCN, and additional sensitivity analyses. RESULTS: DFMO after completion of immunotherapy was associated with improved EFS (hazard ratio [HR], 0.50 [95% CI, 0.29 to 0.84]; P = .008) and OS (HR, 0.38 [95% CI, 0.19 to 0.76]; P = .007). The results were confirmed with propensity score-matched cohorts and sensitivity analyses. CONCLUSION: The externally controlled analyses presented show a relapse risk reduction in patients with HRNB treated with postimmunotherapy DFMO.


Asunto(s)
Eflornitina , Neuroblastoma , Niño , Humanos , Eflornitina/efectos adversos , Puntaje de Propensión , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neuroblastoma/tratamiento farmacológico , Recurrencia , Supervivencia sin Enfermedad
2.
Genome Med ; 16(1): 28, 2024 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-38347552

RESUMEN

BACKGROUND: Children with relapsed central nervous system (CNS tumors), neuroblastoma, sarcomas, and other rare solid tumors face poor outcomes. This prospective clinical trial examined the feasibility of combining genomic and transcriptomic profiling of tumor samples with a molecular tumor board (MTB) approach to make real­time treatment decisions for children with relapsed/refractory solid tumors. METHODS: Subjects were divided into three strata: stratum 1-relapsed/refractory neuroblastoma; stratum 2-relapsed/refractory CNS tumors; and stratum 3-relapsed/refractory rare solid tumors. Tumor samples were sent for tumor/normal whole-exome (WES) and tumor whole-transcriptome (WTS) sequencing, and the genomic data were used in a multi-institutional MTB to make real­time treatment decisions. The MTB recommended plan allowed for a combination of up to 4 agents. Feasibility was measured by time to completion of genomic sequencing, MTB review and initiation of treatment. Response was assessed after every two cycles using Response Evaluation Criteria in Solid Tumors (RECIST). Patient clinical benefit was calculated by the sum of the CR, PR, SD, and NED subjects divided by the sum of complete response (CR), partial response (PR), stable disease (SD), no evidence of disease (NED), and progressive disease (PD) subjects. Grade 3 and higher related and unexpected adverse events (AEs) were tabulated for safety evaluation. RESULTS: A total of 186 eligible patients were enrolled with 144 evaluable for safety and 124 evaluable for response. The average number of days from biopsy to initiation of the MTB-recommended combination therapy was 38 days. Patient benefit was exhibited in 65% of all subjects, 67% of neuroblastoma subjects, 73% of CNS tumor subjects, and 60% of rare tumor subjects. There was little associated toxicity above that expected for the MGT drugs used during this trial, suggestive of the safety of utilizing this method of selecting combination targeted therapy. CONCLUSIONS: This trial demonstrated the feasibility, safety, and efficacy of a comprehensive sequencing model to guide personalized therapy for patients with any relapsed/refractory solid malignancy. Personalized therapy was well tolerated, and the clinical benefit rate of 65% in these heavily pretreated populations suggests that this treatment strategy could be an effective option for relapsed and refractory pediatric cancers. TRIAL REGISTRATION: ClinicalTrials.gov, NCT02162732. Prospectively registered on June 11, 2014.


Asunto(s)
Neuroblastoma , Niño , Humanos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/genética , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/etiología
3.
Brain Sci ; 13(11)2023 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-38002476

RESUMEN

Primitive neuroectodermal tumors of the central nervous system, or CNS neuroblastoma, are rare neoplasms in children. Recently, methylation profiling enabled the discovery of four distinct entities of these tumors. The current treatment paradigm involves surgical resection followed by chemotherapy and radiation. However, upfront surgical resection carries high surgical morbidity in this patient population due to their young age, tumor vascularity, and often deep location in the brain. We report a case of CNS neuroblastoma that can be successfully treated with neoadjuvant chemotherapy followed by minimally invasive laser interstitial thermal therapy and radiation. The patient has complete treatment with no evidence of recurrence at one year follow-up. This case illustrates a potential paradigm shift in the treatment of these rare tumors can be treated using minimally invasive surgical approach to achieve a favorable outcome.

4.
J Clin Oncol ; 41(10): 1921-1932, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36548930

RESUMEN

PURPOSE: Infant and young childhood medulloblastoma (iMB) is usually treated without craniospinal irradiation (CSI) to avoid neurocognitive late effects. Unfortunately, many children relapse. The purpose of this study was to assess salvage strategies and prognostic features of patients with iMB who relapse after CSI-sparing therapy. METHODS: We assembled a large international cohort of 380 patients with relapsed iMB, age younger than 6 years, and initially treated without CSI. Univariable and multivariable Cox models of postrelapse survival (PRS) were conducted for those treated with curative intent using propensity score analyses to account for confounding factors. RESULTS: The 3-year PRS, for 294 patients treated with curative intent, was 52.4% (95% CI, 46.4 to 58.3) with a median time to relapse from diagnosis of 11 months. Molecular subgrouping was available for 150 patients treated with curative intent, and 3-year PRS for sonic hedgehog (SHH), group 4, and group 3 were 60%, 84%, and 18% (P = .0187), respectively. In multivariable analysis, localized relapse (P = .0073), SHH molecular subgroup (P = .0103), CSI use after relapse (P = .0161), and age ≥ 36 months at initial diagnosis (P = .0494) were associated with improved survival. Most patients (73%) received salvage CSI, and although salvage chemotherapy was not significant in multivariable analysis, its use might be beneficial for a subset of children receiving salvage CSI < 35 Gy (P = .007). CONCLUSION: A substantial proportion of patients with relapsed iMB are salvaged after initial CSI-sparing approaches. Patients with SHH subgroup, localized relapse, older age at initial diagnosis, and those receiving salvage CSI show improved PRS. Future prospective studies should investigate optimal CSI doses and the role of salvage chemotherapy in this population.


Asunto(s)
Neoplasias Encefálicas , Neoplasias Cerebelosas , Irradiación Craneoespinal , Meduloblastoma , Niño , Humanos , Lactante , Preescolar , Meduloblastoma/radioterapia , Estudios de Cohortes , Estudios Prospectivos , Irradiación Craneoespinal/efectos adversos , Proteínas Hedgehog , Recurrencia Local de Neoplasia , Neoplasias Encefálicas/terapia , Enfermedad Crónica , Neoplasias Cerebelosas/radioterapia
5.
Cancer Rep (Hoboken) ; 5(11): e1616, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35355452

RESUMEN

BACKGROUND: Survival for patients with high-risk neuroblastoma (HRNB) remains poor despite aggressive multimodal therapies. AIMS: To study the feasibility and safety of incorporating a genomic-based targeted agent to induction therapy for HRNB as well as the feasibility and safety of adding difluoromethylornithine (DFMO) to anti-GD2 immunotherapy. METHODS: Twenty newly diagnosed HRNB patients were treated on this multicenter pilot trial. Molecular tumor boards selected one of six targeted agents based on tumor-normal whole exome sequencing and tumor RNA-sequencing results. Treatment followed standard upfront HRNB chemotherapy with the addition of the selected targeted agent to cycles 3-6 of induction. Following consolidation, DFMO (750 mg/m2 twice daily) was added to maintenance with dinutuximab and isotretinoin, followed by continuation of DFMO alone for 2 years. DNA methylation analysis was performed retrospectively and compared to RNA expression. RESULTS: Of the 20 subjects enrolled, 19 started targeted therapy during cycle 3 and 1 started during cycle 5. Eighty-five percent of subjects met feasibility criteria (receiving 75% of targeted agent doses). Addition of targeted agents did not result in toxicities requiring dose reduction of chemotherapy or permanent discontinuation of targeted agent. Following standard consolidation, 15 subjects continued onto immunotherapy with DFMO. This combination was well-tolerated and resulted in no unexpected adverse events related to DFMO. CONCLUSION: This study demonstrates the safety and feasibility of adding targeted agents to standard induction therapy and adding DFMO to immunotherapy for HRNB. This treatment regimen has been expanded to a Phase II trial to evaluate efficacy.


Asunto(s)
Antineoplásicos , Neuroblastoma , Humanos , Eflornitina/efectos adversos , Proyectos Piloto , Quimioterapia de Inducción , Estudios Retrospectivos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/genética , Inmunoterapia , Antineoplásicos/uso terapéutico , Factores Inmunológicos , Genómica , ARN/uso terapéutico
6.
Cancer Res ; 81(23): 5818-5832, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34610968

RESUMEN

Children with treatment-refractory or relapsed (R/R) tumors face poor prognoses. As the genomic underpinnings driving R/R disease are not well defined, we describe here the genomic and transcriptomic landscapes of R/R solid tumors from 202 patients enrolled in Beat Childhood Cancer Consortium clinical trials. Tumor mutational burden (TMB) was elevated relative to untreated tumors at diagnosis, with one-third of tumors classified as having a pediatric high TMB. Prior chemotherapy exposure influenced the mutational landscape of these R/R tumors, with more than 40% of tumors demonstrating mutational signatures associated with platinum or temozolomide chemotherapy and two tumors showing treatment-associated hypermutation. Immunogenomic profiling found a heterogenous pattern of neoantigen and MHC class I expression and a general absence of immune infiltration. Transcriptional analysis and functional gene set enrichment analysis identified cross-pathology clusters associated with development, immune signaling, and cellular signaling pathways. While the landscapes of these R/R tumors reflected those of their corresponding untreated tumors at diagnosis, important exceptions were observed, suggestive of tumor evolution, treatment resistance mechanisms, and mutagenic etiologies of treatment. SIGNIFICANCE: Tumor heterogeneity, chemotherapy exposure, and tumor evolution contribute to the molecular profiles and increased mutational burden that occur in treatment-refractory and relapsed childhood solid tumors.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/genética , Resistencia a Antineoplásicos , Evasión Inmune , Mutación , Recurrencia Local de Neoplasia/patología , Neoplasias/patología , Adolescente , Adulto , Niño , Preescolar , Femenino , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica , Humanos , Lactante , Estudios Longitudinales , Masculino , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/inmunología , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/inmunología , Pronóstico , Tasa de Supervivencia , Transcriptoma , Adulto Joven
7.
J Pediatr Hematol Oncol ; 32(2): 97-102, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20118811

RESUMEN

Reduction of thrombus size and recanalization of vessels after deep vein thrombosis (DVT) are important goals to prevent recurrent thrombosis and development of postthrombotic syndrome. Thrombolysis is effective but concern for bleeding complications has limited its use in children. We retrospectively analyzed data for children with DVT treated with a low-dose systemic tissue plasminogen activator (tPA) regimen. Twenty-three pediatric patients (12 males and 11 females, median age 12 y) received low-dose systemic tPA, initiated at 0.03 to 0.06 mg/kg/h for a median of 24 hours (range 12 to 48 h). Of the 20 patients imaged within 24 hours of therapy, 6 (30%) showed partial to complete thrombus resolution. Eight patients subsequently received increased tPA at 0.12 mg/kg/h for an additional 24 hours (range 12 to 36 h). Six of these 8 (75%) patients responded to the increased dose. The overall response at the end of thrombolytic therapy was 59% (13/22). Two bleeding complications occurred without serious sequelae. Low-dose tPA administration leads to a substantial response rate although the risk of bleeding remains unclear. A prospective multicenter trial of low-dose thrombolytic therapy in children with acute DVT is warranted.


Asunto(s)
Terapia Trombolítica , Activador de Tejido Plasminógeno/uso terapéutico , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Estudios Retrospectivos , Terapia Trombolítica/efectos adversos , Activador de Tejido Plasminógeno/efectos adversos , Trombosis de la Vena/tratamiento farmacológico
8.
J Neuropathol Exp Neurol ; 78(11): 1011-1021, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31562743

RESUMEN

MAPK pathway activation has been recurrently observed in desmoplastic infantile ganglioglioma/astrocytoma (DIG/DIA) with reported disproportionally low mutation allele frequencies relative to the apparent high tumor content, suggesting that MAPK pathway alterations may be subclonal. We sought to expand the number of molecularly profiled cases and investigate if tumor cell composition could account for the observed low mutation allele frequencies. Molecular (targeted neuro-oncology next-generation sequencing/RNA sequencing and OncoScan microarray) and immunohistochemical (CD68-PGM1/CD163/CD14/CD11c/lysozyme/CD3/CD20/CD34/PD-L1) studies were performed in 7 DIG. Activating MAPK pathway alterations were identified in 4 (57%) cases: 3 had a BRAF mutation (V600E/V600D/V600_W604delinsDQTDG, at 8%-27% variant allele frequency) and 1 showed a TPM3-NRTK1 fusion. Copy number changes were infrequent and nonrecurrent. All tumors had at least 30% of cells morphologically and immunophenotypically consistent with microglial/macrophage lineage. Two subtotally resected tumors regrew; 1 was re-excised and received adjuvant treatment (chemotherapy/targeted therapy), with clinical response to targeted therapy only. Even with residual tumor, all patients are alive (median follow-up, 83 months; 19-139). This study further supports DIG as another MAPK pathway-driven neuroepithelial tumor, thus expanding potential treatment options for tumors not amenable to surgical cure, and suggests that DIG is a microglia/macrophage-rich neuroepithelial tumor with frequent low driver mutation allele frequencies.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Ganglioglioma/metabolismo , Ganglioglioma/patología , Sistema de Señalización de MAP Quinasas , Macrófagos/metabolismo , Microglía/metabolismo , Neoplasias Neuroepiteliales/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/patología , Femenino , Humanos , Lactante , Macrófagos/patología , Masculino , Microglía/patología , Neoplasias Neuroepiteliales/patología
9.
Pediatr Hematol Oncol ; 25(5): 423-9, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18569844

RESUMEN

Hydroxyurea has documented laboratory and clinical efficacy for children with sickle cell anemia (SCA), and has potential to become an effective and inexpensive treatment option for patients in countries with limited resources. Concerns exist, however, regarding product quality and manufacturing variability among different international vendors, particularly for generic formulations. To address these concerns, hydroxyurea capsules from 8 different pharmaceutical sources were analyzed using quantitative chemical and functional assays. All samples had measured values within 20% of expected results, with no significant differences observed among vendors. Generic hydroxyurea formulations represent a potent yet inexpensive therapeutic option for children with SCA worldwide.


Asunto(s)
Medicamentos Genéricos/análisis , Hidroxiurea/análisis , Anemia de Células Falciformes/tratamiento farmacológico , Antidrepanocíticos , Medicamentos Genéricos/normas , Humanos , Hidroxiurea/normas , Preparaciones Farmacéuticas/análisis , Preparaciones Farmacéuticas/normas , Análisis Espectral
10.
Exp Hematol ; 35(2): 179-83, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17258066

RESUMEN

OBJECTIVES: Although functional asplenia in sickle cell disease (SCD) begins early in life and has important clinical consequences, quantitative measurement of splenic function is not readily available. A novel high-throughput flow cytometric method for quantitating Howell-Jolly bodies (HJB) has been developed which isolates HJB-containing CD71(+) and CD71(-) erythrocytes. Analysis of these cell populations allows quantitative measurement of splenic filtrative function and possible chromosomal damage. METHODS: Blood specimens from 147 children with SCD were analyzed using a high-throughput flow cytometric method. Enumeration of the following populations was accomplished: 1) CD71(+) reticulocytes among total erythrocytes, identifying the youngest erythroid cell population; 2) HJB-containing CD71(+) reticulocytes, which isolate young erythrocytes containing micronuclei as an index of cytogenetic damage; and 3) HJB-containing CD71(-) erythrocytes, identifying older erythrocytes containing micronuclei, indirectly measuring splenic function. RESULTS: Children with HbSC (n = 24) had slightly elevated HJB frequencies, while children with HbSS (n = 125) had highly elevated frequencies within CD71(+) cells (0.44% +/- 0.40%, normal 0.12% +/- 0.06%, p < 0.001) and CD71(-) cells (2493 +/- 2303 per million RBC, normal 20 +/- 11, p < 0.001). Using a multiple regression model, the frequency of HbSS CD71(+) reticulocytes containing HJB was significantly influenced by hydroxyurea use (p < 0.0001), age (p = 0.0288), and splenectomy (p = 0.0498). Similarly, mature CD71(-) erythrocytes containing HJB were positively correlated with hydroxyurea (p = 0.0001), age (p < 0.0001), and splenectomy (p = 0.0104). CONCLUSIONS: HJB quantitation by flow cytometry is a novel assay for measuring splenic function and may be valuable for investigating the efficacy and safety of therapeutic options for children with SCD.


Asunto(s)
Anemia de Células Falciformes/sangre , Inclusiones Eritrocíticas/patología , Citometría de Flujo/métodos , Adolescente , Adulto , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/metabolismo , Antígenos CD/metabolismo , Niño , Preescolar , Estudios de Cohortes , Inclusiones Eritrocíticas/metabolismo , Eritrocitos/metabolismo , Eritrocitos/patología , Femenino , Genotipo , Hemoglobina Falciforme/análisis , Humanos , Lactante , Recién Nacido , Masculino , Pruebas de Micronúcleos , Receptores de Transferrina/metabolismo , Reproducibilidad de los Resultados , Reticulocitos/metabolismo , Reticulocitos/patología , Esplenectomía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA