Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
J Glob Antimicrob Resist ; 22: 354-357, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32251868

RESUMEN

OBJECTIVES: In the pre-azole era, central nervous system (CNS) infections with Aspergillus had a dismal outcome. Survival improved with voriconazole but CNS infections caused by azole-resistant Aspergillus fumigatus preclude its use. Intravenous liposomal-amphotericin B (L-AmB) is the preferred treatment option for azole-resistant CNS infections but has suboptimal brain concentrations. METHODS: We describe three patients with biopsy-proven CNS aspergillosis where intraventricular L-AmB was added to systemic therapy. Two patients with azole-resistant aspergillosis and one patient with azole-susceptible CNS aspergillosis were treated with intraventricular L-AmB at a dose of 1mg weekly. RESULTS: We describe three patients successfully treated with a combination of intravenous and intraventricular L-AmB. All three patients survived but one patient developed serious headaches, most likely not related to this treatment. CONCLUSIONS: Intraventricular L-AmB may have a role in the treatment of therapy-refractory CNS aspergillosis when added to systemic therapy.


Asunto(s)
Anfotericina B , Aspergillus fumigatus , Anfotericina B/farmacología , Anfotericina B/uso terapéutico , Antifúngicos/farmacología , Antifúngicos/uso terapéutico , Azoles/farmacología , Azoles/uso terapéutico , Farmacorresistencia Fúngica , Humanos
2.
Clin Pharmacokinet ; 59(2): 207-216, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31313068

RESUMEN

INTRODUCTION: The pharmacokinetics (PK) of the 20S proteasome inhibitor bortezomib are characterized by a large volume of distribution and a rapid decline in plasma concentrations within the first hour after administration. An increase in exposure was observed in the second week of treatment, which has previously been explained by extensive binding of bortezomib to proteasome in erythrocytes and peripheral tissues. We characterized the nonlinear population PK and pharmacodynamics (PD) of bortezomib in children with acute lymphoblastic leukemia. METHODS: Overall, 323 samples from 28 patients were available from a pediatric clinical study investigating bortezomib at an intravenous dose of 1.3 mg/m2 twice weekly (Dutch Trial Registry number 1881/ITCC021). A semi-physiological PK model for bortezomib was first developed; the PK were linked to the decrease in 20S proteasome activity in the final PK/PD model. RESULTS: The plasma PK data were adequately described using a two-compartment model with linear elimination. Increased concentrations were observed in week 2 compared with week 1, which was described using a Langmuir binding model. The decrease in 20S proteasome activity was best described by a direct effect model with a sigmoidal maximal inhibitory effect, representing the relationship between plasma concentrations and effect. The maximal inhibitory effect was 0.696 pmol AMC/s/mg protein (95% confidence interval 0.664-0.728) after administration. CONCLUSION: The semi-physiological model adequately described the nonlinear PK and PD of bortezomib in plasma. This model can be used to further optimize dosing of bortezomib.


Asunto(s)
Bortezomib/farmacocinética , Eritrocitos/efectos de los fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Inhibidores de Proteasoma/farmacocinética , Administración Intravenosa , Adolescente , Bortezomib/administración & dosificación , Bortezomib/sangre , Bortezomib/uso terapéutico , Niño , Preescolar , Relación Dosis-Respuesta a Droga , Eritrocitos/metabolismo , Estudios de Factibilidad , Femenino , Humanos , Lactante , Masculino , Modelos Biológicos , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Inhibidores de Proteasoma/administración & dosificación , Inhibidores de Proteasoma/sangre , Inhibidores de Proteasoma/uso terapéutico , Recurrencia
3.
Clin Infect Dis ; 71(8): e281-e288, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31790556

RESUMEN

BACKGROUND: Ciprofloxacin is used as antimicrobial prophylaxis in pediatric acute lymphoblastic leukemia (ALL) to decrease infections with gram-negative bacteria. However, there are no clear guidelines concerning prophylactic dose. AIMS: To determine the pharmacokinetics and pharmacodynamics (PKPD) of ciprofloxacin prophylaxis in a pediatric ALL population. The effect of patient characteristics and antileukemic treatment on ciprofloxacin exposure, the area under the concentration time curve over minimal inhibitory concentration (AUC24/MIC) ratios, and emergence of resistance were studied. METHODS: A total of 615 samples from 129 children (0-18 years) with ALL were collected in a multicenter prospective study. A population pharmacokinetic model was developed. Microbiological cultures were collected prior to and during prophylaxis. An AUC24/MIC of ≥125 was defined as target ratio. RESULTS: A 1-compartment model with zero-order absorption and allometric scaling best described the data. No significant (P < .01) covariates remained after backward elimination and no effect of asparaginase or azoles were found. Ciprofloxacin AUC24 was 16.9 mg*h/L in the prednisone prophase versus 29.3 mg*h/L with concomitant chemotherapy. Overall, 100%, 81%, and 18% of patients at, respectively, MIC of 0.063, 0.125, and 0.25 mg/L achieved AUC24/MIC ≥ 125. In 13% of the patients, resistant bacteria were found during prophylactic treatment. CONCLUSION: Ciprofloxacin exposure shows an almost 2-fold change throughout the treatment of pediatric ALL. Depending on the appropriateness of 125 as target ratio, therapeutic drug monitoring or dose adjustments might be indicated for less susceptible bacteria starting from ≥ 0.125 mg/L to prevent the emergence of resistance and reach required targets for efficacy.


Asunto(s)
Ciprofloxacina , Leucemia-Linfoma Linfoblástico de Células Precursoras , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Área Bajo la Curva , Niño , Ciprofloxacina/farmacología , Ciprofloxacina/uso terapéutico , Farmacorresistencia Bacteriana , Humanos , Pruebas de Sensibilidad Microbiana , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Estudios Prospectivos
4.
Leukemia ; 33(6): 1349-1359, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30568171

RESUMEN

Variation in survival of pediatric acute myeloid leukemia (pAML) over time and between Western European countries exists. The aim of the current study is to assess the progress made for the Dutch pAML population (0-17 years) during 1990-2015, based on trends in incidence, survival and mortality. Data from the population-based Netherlands Cancer Registry were merged with leukemia-related characteristics and treatment specifics from the Dutch Childhood Leukemia Study Group (Dutch Childhood Oncology Group (DCOG) from 2002 onwards). Mortality data (1980-2016) were obtained from the cause of death registry of Statistics Netherlands. Trend analyses were performed over time and by treatment protocol. Between 1990 and 2015, a total of 635 children aged 0-17 years were diagnosed with AML for an average of 25 patients (range 18-36) per year. There was a slight increase in the incidence at age 1-4 years (average annual percentage change (AAPC) of +2.2% per year (95% CI 0.8-3.5, p < 0.01)). Overall, the 5-year survival significantly improved over the past 26 years and nearly doubled from 40% in the early 1990s to 74% in 2010-2015. Multivariable analysis showed a 49% reduction in risk of death for pAML patients treated according to the latest DB-AML 01 protocol (p = 0.03). The continuing decrease of mortality (AAPC -2.8% per year (95% CI -4.1 to -1.5)) supports the conclusion of true progress against pAML in the Netherlands.


Asunto(s)
Leucemia Mieloide Aguda/epidemiología , Leucemia Mieloide Aguda/mortalidad , Mortalidad/tendencias , Sistema de Registros/estadística & datos numéricos , Adolescente , Adulto , Niño , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Incidencia , Lactante , Recién Nacido , Leucemia Mieloide Aguda/patología , Masculino , Países Bajos/epidemiología , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia , Adulto Joven
5.
Oncogene ; 37(1): 107-115, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-28892045

RESUMEN

In acute myeloid leukemia (AML), specific genomic aberrations induce aberrant methylation, thus directly influencing the transcriptional programing of leukemic cells. Therefore, therapies targeting epigenetic processes are advocated as a promising therapeutic tool for AML treatment. However, to develop new therapies, a comprehensive understanding of the mechanism(s) driving the epigenetic changes as a result of acquired genetic abnormalities is necessary. This understanding is still lacking. In this study, we performed genome-wide CpG-island methylation profiling on pediatric AML samples. Six differentially methylated genomic regions within two genes, discriminating inv(16)(p13;q22) from non-inv(16) pediatric AML samples, were identified. All six regions had a hypomethylated phenotype in inv(16) AML samples, and this was most prominent at the regions encompassing the meningioma (disrupted in balanced translocation) 1 (MN1) oncogene. MN1 expression primarily correlated with the methylation level of the 3' end of the MN1 exon-1 locus. Decitabine treatment of different cell lines showed that induced loss of methylation at the MN1 locus can result in an increase of MN1 expression, indicating that MN1 expression is coregulated by DNA methylation. To investigate this methylation-associated mechanism, we determined the expression of DNA methyltransferases in inv(16) AML. We found that DNMT3B expression was significantly lower in inv(16) samples. Furthermore, DNMT3B expression correlated negatively with MN1 expression in pediatric AML samples. Importantly, depletion of DNMT3B impaired remethylation efficiency of the MN1 exon-1 locus in AML cells after decitabine exposure. These findings identify DNMT3B as an important coregulator of MN1 methylation. Taken together, this study shows that the methylation level of the MN1 exon-1 locus regulates MN1 expression levels in inv(16) pediatric AML. This methylation level is dependent on DNMT3B, thus suggesting a role for DNMT3B in leukemogenesis in inv(16) AML, through MN1 methylation regulation.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/genética , Proteínas Supresoras de Tumor/genética , Adolescente , Azacitidina/análogos & derivados , Azacitidina/farmacología , Carcinogénesis/genética , Línea Celular Tumoral , Niño , Preescolar , Islas de CpG/genética , Metilación de ADN/efectos de los fármacos , Decitabina , Epigénesis Genética/genética , Exones/genética , Femenino , Humanos , Lactante , Recién Nacido , Leucemia Mieloide Aguda/sangre , Leucemia Mieloide Aguda/patología , Masculino , Hibridación de Ácido Nucleico/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Proteínas de Fusión Oncogénica/genética , Regiones Promotoras Genéticas/genética , Transactivadores , ADN Metiltransferasa 3B
6.
Leukemia ; 32(3): 828-836, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28871137

RESUMEN

Overexpression of the BRE (brain and reproductive organ-expressed) gene defines a distinct pediatric and adult acute myeloid leukemia (AML) subgroup. Here we identify a promoter enriched for active chromatin marks in BRE intron 4 causing strong biallelic expression of a previously unknown C-terminal BRE transcript. This transcript starts with BRE intron 4 sequences spliced to exon 5 and downstream sequences, and if translated might code for an N terminally truncated BRE protein. Remarkably, the new BRE transcript was highly expressed in over 50% of 11q23/KMT2A (lysine methyl transferase 2A)-rearranged and t(8;16)/KAT6A-CREBBP cases, while it was virtually absent from other AML subsets and normal tissues. In gene reporter assays, the leukemia-specific fusion protein KMT2A-MLLT3 transactivated the intragenic BRE promoter. Further epigenome analyses revealed 97 additional intragenic promoter marks frequently bound by KMT2A in AML with C-terminal BRE expression. The corresponding genes may be part of a context-dependent KMT2A-MLLT3-driven oncogenic program, because they were higher expressed in this AML subtype compared with other groups. C-terminal BRE might be an important contributor to this program because in a case with relapsed AML, we observed an ins(11;2) fusing CHORDC1 to BRE at the region where intragenic transcription starts in KMT2A-rearranged and KAT6A-CREBBP AML.


Asunto(s)
Reordenamiento Génico , Leucemia Mieloide Aguda/genética , Proteínas del Tejido Nervioso/genética , Dominios y Motivos de Interacción de Proteínas/genética , Activación Transcripcional , Translocación Genética , Línea Celular , Cromosomas Humanos Par 11 , Cromosomas Humanos Par 16 , Epigénesis Genética , Exones , Regulación Leucémica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Humanos , Intrones , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas Nucleares/genética , Regiones Promotoras Genéticas
7.
Eur J Cancer ; 90: 102-110, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29274926

RESUMEN

INTRODUCTION: Prognosis of neuroblastoma patients is very diverse, indicating the need for more accurate prognostic parameters. The excretion of catecholamine metabolites by most neuroblastomas is used for diagnostic purposes, but their correlation with prognosis has hardly been investigated. Therefore, we performed an in-depth analysis of a panel of elevated urinary catecholamine metabolites at diagnosis and their correlation with prognosis. PATIENTS AND METHODS: Retrospective study of eight urinary catecholamine metabolites in a test (n = 96) and validation (n = 205) cohort of patients with neuroblastoma (all stages) at diagnosis. RESULTS: Multivariate analyses, including risk factors such as stage and MYCN amplification, revealed that 3-methoxytyramine (3MT) was an independent risk factor for event-free survival (EFS) and overall survival (OS). Furthermore, only 3MT appeared to be an independent risk factor for both EFS and OS in high-risk patients, which was independent of modern high-risk therapy and immunotherapy. Among high-risk patients, those with elevated 3MT and older than 18 months had an extremely poor prognosis compared to patients with non-elevated 3MT and younger than 18 months (5-year EFS of 14.3% ± 4% and 66.7% ± 18%, respectively, p = 0.001; 5-year OS of 21.8% ± 5% and 87.5% ± 12%, respectively, p < 0.001). CONCLUSIONS: Elevated 3MT at diagnosis was associated with high-risk disease and poor prognosis. For high-risk patients, elevated 3MT at diagnosis was the only significant risk factor for EFS and OS. 3MT was also able to identify subgroups of high-risk patients with favourable and extremely poor prognosis.


Asunto(s)
Biomarcadores de Tumor/orina , Dopamina/análogos & derivados , Neuroblastoma/patología , Adolescente , Niño , Preescolar , Estudios de Cohortes , Supervivencia sin Enfermedad , Dopamina/orina , Femenino , Humanos , Lactante , Recién Nacido , Estimación de Kaplan-Meier , Masculino , Neuroblastoma/mortalidad , Neuroblastoma/orina , Pronóstico , Estudios Retrospectivos , Resultado del Tratamiento
8.
Leukemia ; 32(4): 882-889, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29089643

RESUMEN

Despite advances in our understanding of the molecular basis for particular subtypes of acute myeloid leukemia (AML), effective therapy remains a challenge for many individuals suffering from this disease. A significant proportion of both pediatric and adult AML patients cannot be cured and since the upper limits of chemotherapy intensification have been reached, there is an urgent need for novel therapeutic approaches. The transcription factor c-MYB has been shown to play a central role in the development and progression of AML driven by several different oncogenes, including mixed lineage leukemia (MLL)-fusion genes. Here, we have used a c-MYB gene expression signature from MLL-rearranged AML to probe the Connectivity Map database and identified mebendazole as a c-MYB targeting drug. Mebendazole induces c-MYB degradation via the proteasome by interfering with the heat shock protein 70 (HSP70) chaperone system. Transient exposure to mebendazole is sufficient to inhibit colony formation by AML cells, but not normal cord blood-derived cells. Furthermore, mebendazole is effective at impairing AML progression in vivo in mouse xenotransplantation experiments. In the context of widespread human use of mebendazole, our data indicate that mebendazole-induced c-MYB degradation represents a safe and novel therapeutic approach for AML.


Asunto(s)
Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Mebendazol/farmacología , Proteolisis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myb/metabolismo , Animales , Niño , Femenino , Humanos , Lactante , Masculino , Ratones , Oncogenes/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo
9.
Leukemia ; 32(4): 931-940, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28972594

RESUMEN

RAS pathway mutations have been linked to relapse and chemotherapy resistance in pediatric B-cell precursor acute lymphoblastic leukemia (BCP-ALL). However, comprehensive data on the frequency and prognostic value of subclonal mutations in well-defined subgroups using highly sensitive and quantitative methods are lacking. Targeted deep sequencing of 13 RAS pathway genes was performed in 461 pediatric BCP-ALL cases at initial diagnosis and in 19 diagnosis-relapse pairs. Mutations were present in 44.2% of patients, with 24.1% carrying a clonal mutation. Mutation frequencies were highest in high hyperdiploid, infant t(4;11)-rearranged, BCR-ABL1-like and B-other cases (50-70%), whereas mutations were less frequent in ETV6-RUNX1-rearranged, and rare in TCF3-PBX1- and BCR-ABL1-rearranged cases (27-4%). RAS pathway-mutated cells were more resistant to prednisolone and vincristine ex vivo. Clonal, but not subclonal, mutations were linked to unfavorable outcome in standard- and high-risk-treated patients. At relapse, most RAS pathway mutations were clonal (9 of 10). RAS mutant cells were sensitive to the MEK inhibitor trametinib ex vivo, and trametinib sensitized resistant cells to prednisolone. We conclude that RAS pathway mutations are frequent, and that clonal, but not subclonal, mutations are associated with unfavorable risk parameters in newly diagnosed pediatric BCP-ALL. These mutations may designate patients eligible for MEK inhibitor treatment.


Asunto(s)
Linfocitos B/metabolismo , Biomarcadores de Tumor/genética , Mutación/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Proteínas ras/genética , Adolescente , Animales , Línea Celular Tumoral , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Ratones , Ratones Endogámicos NOD , Tasa de Mutación , Proteínas de Fusión Oncogénica/genética , Pronóstico , Transducción de Señal/genética
10.
Neth J Med ; 75(10): 428-431, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29256412

RESUMEN

The protein kinase inhibitor dasatinib, targeting BCR-ABL and Src family kinases, is used in chronic myeloid leukaemia and Philadelphia-chromosome positive acute lymphoblastic leukaemia. The Netherlands Pharmacovigilance Centre Lareb has received one report of nephrotic syndrome associated with the use of dasatinib. With some other protein kinase inhibitors, targeting vascular endothelial growth factor, nephrotic syndrome is a well-known adverse drug reaction. The Dutch and European pharmacovigilance databases and scientific literature contain several cases indicating a causal relationship between dasatinib and nephrotic syndrome. Nephrotic syndrome was recently added to the list of adverse drug reactions in the Dutch summary of product characteristics for dasatinib. It is important to recognise the possibility of this adverse drug reaction when a patient develops nephrotic syndrome under treatment with dasatinib.


Asunto(s)
Antineoplásicos/efectos adversos , Dasatinib/efectos adversos , Síndrome Nefrótico/inducido químicamente , Inhibidores de Proteínas Quinasas/efectos adversos , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Lactante , Leucemia Mieloide/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Proteinuria/inducido químicamente
12.
Pediatr Blood Cancer ; 64(3)2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27667142

RESUMEN

Transient myeloproliferative disorder (TMD) is a leukemia type that occurs typically in newborns. In Down syndrome, TMD is referred to as transient abnormal myelopoiesis (TAM).32 Recently, transientness has also been reported in acute myeloid leukemia patients with germline trisomy 21 mosaicism, and even in cases with somatic trisomy 21, with or without GATA1 mutations. TMD cases without trisomy 21 are rare, and recurrent genetic aberrations that aid in clinical decision-making are scarcely described. We describe here a TMD patient without trisomy 21 or GATA1 mutation in whom single-nucleotide polymorphism analysis of leukemic blasts revealed a novel combined submicroscopic deletion (5q31.1-5q31.3 and 8q23.2q24).


Asunto(s)
Cromosomas Humanos Par 5/genética , Cromosomas Humanos Par 8/genética , Síndrome de Down/genética , Factor de Transcripción GATA1/genética , Leucemia Megacarioblástica Aguda/genética , Polimorfismo de Nucleótido Simple/genética , Síndrome de Down/patología , Humanos , Recién Nacido , Leucemia Megacarioblástica Aguda/patología , Pronóstico
13.
Expert Rev Anticancer Ther ; 16(2): 137-46, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26646091

RESUMEN

Gemtuzumab ozogamicin, an anti-tumour antibiotic linked to an anti-CD33 antibody (Mylotarg®), has been well studied in AML in adults but to a lesser extent in children. No review has yet been published on the dose-related toxicity and efficacy of gemtuzumab ozogamicin in pediatric AML patients. Here we looked at 14 studies then scatterplots and linear regressions were used to estimate the relationship between the dose of gemtuzumab and its toxicity and efficacy. A non-significant increase in bilirubin level and in incidence of veno-occlusive disease was seen with higher doses of gemtuzumab ozogamicin when used as single-agent. In terms of efficacy, even a low dose of 3 mg/m(2) of gemtuzumab ozogamicin can have antileukemic effect, but available data do not allow conclusions on its dose-dependency. Data indicate that higher doses of gemtuzumab ozogamicin account for more adverse events. The data do not show that a high dose is required for anti-leukemic efficacy of gemtuzumab ozogamicin. This study also indicates that there seems to be a role for gemtuzumab ozogamicin in the treatment of pediatric AML and further studies are required to assess its optimal dose, schedule and balance between efficacy and side-effects.


Asunto(s)
Aminoglicósidos/administración & dosificación , Anticuerpos Monoclonales Humanizados/administración & dosificación , Antineoplásicos/administración & dosificación , Leucemia Mieloide Aguda/tratamiento farmacológico , Factores de Edad , Aminoglicósidos/efectos adversos , Aminoglicósidos/uso terapéutico , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Niño , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Gemtuzumab , Humanos , Leucemia Mieloide Aguda/patología , Modelos Lineales
14.
Mol Cancer Res ; 13(6): 982-92, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25944917

RESUMEN

UNLABELLED: Loss of ephrin receptor (EphB1) expression may associate with aggressive cancer phenotypes; however, the mechanism of action remains unclear. To gain detailed insight into EphB1 function in acute myelogenous leukemia (AML), comprehensive analysis of EphB1 transcriptional regulation was conducted. In AML cells, EphB1 transcript was inversely correlated with EphB1 promoter methylation. The presence of EphB1 allowed EfnB1 ligand-mediated p53 DNA binding, leading to restoration of the DNA damage response (DDR) cascade by the activation of ATR, Chk1, p53, p21, p38, CDK1(tyr15), and Bax, and downregulation of HSP27 and Bcl2. Comparatively, reintroduction of EphB1 expression in EphB1-methylated AML cells enhanced the same cascade of ATR, Chk1, p21, and CDK1(tyr15), which consequently enforced programmed cell death. Interestingly, in pediatric AML samples, EphB1 peptide phosphorylation and mRNA expression were actively suppressed as compared with normal bone marrow, and a significant percentage of the primary AML specimens had EphB1 promoter hypermethylation. Finally, EphB1 repression associated with a poor overall survival in pediatric AML. Combined, the contribution of EphB1 to the DDR system reveals a tumor-suppressor function for EphB1 in pediatric AML. IMPLICATIONS: The tumor-suppressor function of EphB1 is clinically relevant across many malignancies, suggesting that EphB1 is an important regulator of common cancer cell transforming pathways.


Asunto(s)
Daño del ADN , Regulación hacia Abajo , Leucemia Mieloide Aguda/metabolismo , Receptor EphB2/metabolismo , Apoptosis , Médula Ósea , Línea Celular Tumoral , Niño , Metilación de ADN , Reparación del ADN , Puntos de Control de la Fase G2 del Ciclo Celular , Humanos , Leucemia Mieloide Aguda/patología , Regiones Promotoras Genéticas , Receptor EphA1/metabolismo
15.
Blood Cancer J ; 4: e209, 2014 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-24786393

RESUMEN

Immunosuppressive therapy (IST), consisting of antithymocyte globulin and cyclosporine A, is effective in refractory cytopenia of childhood (RCC), suggesting that, similar to low-grade myelodysplastic syndromes in adult patients, T lymphocytes are involved in suppressing hematopoiesis in a subset of RCC patients. However, the potential role of a T-cell-mediated pathophysiology in RCC remains poorly explored. In a cohort of 92 RCC patients, we prospectively assessed the frequency of T-cell receptor (TCR) ß-chain variable (Vß) domain skewing in bone marrow and peripheral blood by heteroduplex PCR, and analyzed T-cell subsets in peripheral blood by flow cytometry. TCRVß skewing was present in 40% of RCC patients. TCRVß skewing did not correlate with bone marrow cellularity, karyotype, transfusion history, HLA-DR15 or the presence of a PNH clone. In 28 patients treated with IST, TCRVß skewing was not clearly related with treatment response. However, TCRVß skewing did correlate with a disturbed CD4(+)/CD8(+) T-cell ratio, a reduction in naive CD8(+) T cells, an expansion of effector CD8(+) T cells and an increase in activated CD8(+) T cells (defined as HLA-DR(+), CD57(+) or CD56(+)). These data suggest that T lymphocytes contribute to RCC pathogenesis in a proportion of patients, and provide a rationale for treatment with IST in selected patients with RCC.


Asunto(s)
Síndromes Mielodisplásicos/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Subgrupos de Linfocitos T/química , Linfocitos T Citotóxicos/inmunología , Adolescente , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Humanos , Terapia de Inmunosupresión , Lactante , Masculino , Síndromes Mielodisplásicos/patología , Pancitopenia/inmunología , Estudios Prospectivos
17.
Leukemia ; 28(5): 1022-32, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24270738

RESUMEN

MicroRNAs (miRNAs) play a pivotal role in the regulation of hematopoiesis and development of leukemia. Great interest emerged in modulating miRNA expression for therapeutic purposes. In order to identify miRNAs, which specifically suppress leukemic growth of acute myeloid leukemia (AML) with t(8;21), inv(16) or mixed lineage leukemia (MLL) rearrangement by inducing differentiation, we conducted a miRNA expression profiling in a cohort of 90 cytogenetically characterized, de novo pediatric AML cases. Four miRNAs, specifically downregulated in MLL-rearranged, t(8;21) or inv(16) AMLs, were characterized by their tumor-suppressive properties in cell lines representing those respective cytogenetic groups. Among those, forced expression of miR-9 reduced leukemic growth and induced monocytic differentiation of t(8;21) AML cell lines in vitro and in vivo. The tumor-suppressive functions of miR-9 were specifically restricted to AML cell lines and primary leukemic blasts with t(8;21). On the other hand, these functions were not evident in AML blasts from patients with MLL rearrangements. We showed that miR-9 exerts its effects through the cooperation with let-7 to repress the oncogenic LIN28B/HMGA2 axis. Thus, miR-9 is a tumor suppressor-miR which acts in a stringent cell context-dependent manner.


Asunto(s)
Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Genes Supresores de Tumor , Leucemia Mieloide Aguda/genética , MicroARNs/genética , Translocación Genética , Animales , División Celular , Niño , Femenino , Citometría de Flujo , Xenoinjertos , Humanos , Leucemia Mieloide Aguda/patología , Masculino , Ratones , Ratones Desnudos
19.
Pediatr Blood Cancer ; 61(6): 1007-12, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24376007

RESUMEN

BACKGROUND: Malignant pleural effusion (MPE) and ascites (MA) negatively impact quality of life of palliative patients. Treatment options are limited. This study's purpose is to examine the experience with indwelling tunneled catheters (ITCs) for management of MPE/MA in children with advanced cancer. METHODS: Children with MPE/MA who underwent ITC insertion (2007-2012) were retrospectively reviewed. Clinical, procedural, complication and outcome details were analyzed. RESULTS: PleurX® ITCs (n = 12) were inserted in eight patients (5-18 years) with sarcoma (11 MPE, 1 MA), achieving symptom relief and facilitating discharge home post ITC (median 2 days). Median survival following ITC was 51 days. There were two major complications: pain (n = 1), late site infection (n = 1), and five minor complications. Drainage ceased in four patients (pleurodesis/tumor progression). At time of death, six ITCs (five patients) were still in situ. CONCLUSIONS: ITC appears to be a safe, effective treatment for MPE/MA in advanced pediatric cancer, achieving symptomatic relief and discharge home.


Asunto(s)
Ascitis/terapia , Catéteres de Permanencia , Drenaje/instrumentación , Derrame Pleural Maligno/terapia , Adolescente , Ascitis/etiología , Ascitis/cirugía , Infecciones Relacionadas con Catéteres/etiología , Catéteres de Permanencia/efectos adversos , Niño , Preescolar , Drenaje/métodos , Femenino , Servicios de Atención de Salud a Domicilio , Humanos , Masculino , Dolor/etiología , Cuidados Paliativos , Paracentesis , Aceptación de la Atención de Salud , Derrame Pleural Maligno/etiología , Derrame Pleural Maligno/cirugía , Pleurodesia , Calidad de Vida , Recurrencia , Estudios Retrospectivos , Sarcoma/complicaciones
20.
Leukemia ; 27(7): 1451-60, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23563239

RESUMEN

Significant improvements in survival for children with acute myeloid leukaemia (AML) have been made over the past three decades, with overall survival rates now approximately 60-70%. However, these gains can be largely attributed to more intensive use of conventional cytotoxics made possible by advances in supportive care, and although over 90% of children achieve remission with frontline therapy, approximately one third in current protocols relapse. Furthermore, late effects of therapy cause significant morbidity for many survivors. Novel therapies are therefore desperately needed. Early-phase paediatric trials of several new agents such as clofarabine, sorafenib and gemtuzumab ozogamicin have shown encouraging results in recent years. Due to the relatively low incidence of AML in childhood, the success of paediatric early-phase clinical trials is largely dependent upon collaborative clinical trial design by international cooperative study groups. Successfully incorporating novel therapies into frontline therapy remains a challenge, but the potential for significant improvement in the duration and quality of survival for children with AML is high.


Asunto(s)
Nucleótidos de Adenina/uso terapéutico , Aminoglicósidos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Arabinonucleósidos/uso terapéutico , Leucemia Monocítica Aguda/tratamiento farmacológico , Niacinamida/análogos & derivados , Compuestos de Fenilurea/uso terapéutico , Antineoplásicos/uso terapéutico , Niño , Clofarabina , Gemtuzumab , Humanos , Inmunotoxinas/uso terapéutico , Niacinamida/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Sorafenib
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...