Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Ann Oncol ; 31(3): 422-429, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32067684

RESUMEN

BACKGROUND: Long-term survival of high-risk neuroblastoma patients is still below 50% despite intensive multimodal treatment. This trial aimed to address whether the addition of two topotecan-containing chemotherapy courses compared to standard induction therapy improves event-free survival (EFS) of these patients. PATIENTS AND METHODS: An open-label, multicenter, prospective randomized controlled trial was carried out at 58 hospitals in Germany and Switzerland. Patients aged 1-21 years with stage 4 neuroblastoma and patients aged 6 months to 21 years with MYCN-amplified tumors were eligible. The primary endpoint was EFS. Patients were randomly assigned to standard induction therapy with six chemotherapy courses or to experimental induction chemotherapy starting with two additional courses of topotecan, cyclophosphamide, and etoposide followed by standard induction chemotherapy (eight courses in total). After induction chemotherapy, all patients received high-dose chemotherapy with autologous hematopoietic stem cell rescue and isotretinoin for consolidation. Radiotherapy was applied to patients with active tumors at the end of induction chemotherapy. RESULTS: Of 536 patients enrolled in the trial, 422 were randomly assigned to the control arm (n = 211) and the experimental arm (n = 211); the median follow-up time was 3.32 years (interquartile range 1.65-5.92). At data lock, the 3-year EFS of experimental and control patients was 34% and 32% [95% confidence Interval (CI) 28% to 40% and 26% to 38%; P = 0.258], respectively. Similarly, the 3-year overall survival of the patients did not differ [54% and 48% (95% CI 46% to 62% and 40% to 56%), respectively; P = 0.558]. The response to induction chemotherapy was not different between the arms. The median number of non-fatal toxicities per patient was higher in the experimental group while the median number of toxicities per chemotherapy course was not different. CONCLUSION: While the burden for the patients was increased by prolonging the induction chemotherapy and the toxicity, the addition of two topotecan-containing chemotherapy courses did not improve the EFS of high-risk neuroblastoma patients and thus cannot be recommended. CLINICAL TRIALS. GOV NUMBER: NCT number 03042429.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Quimioterapia de Inducción , Neuroblastoma , Adolescente , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Niño , Preescolar , Supervivencia sin Enfermedad , Alemania , Humanos , Lactante , Neuroblastoma/tratamiento farmacológico , Estudios Prospectivos , Suiza , Resultado del Tratamiento , Adulto Joven
2.
J Musculoskelet Neuronal Interact ; 16(3): 183-92, 2016 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-27609033

RESUMEN

OBJECTIVES: to investigate feasibility, safety and efficacy of home-based side-alternating whole body vibration (sWBV) to improve motor function in toddlers with cerebral palsy (CP). METHODS: Randomized controlled trial including 24 toddlers with CP (mean age 19 months (SD±3.1); 13 boys). INTERVENTION: 14 weeks sWBV with ten 9-minute sessions weekly (non-individualized). Group A started with sWBV, followed by 14 weeks without; in group B this order was reversed. Feasibility (≥70% adherence) and adverse events were recorded; efficacy evaluated with the Gross Motor Function Measure (GMFM-66), Pediatric Evaluation of Disability Inventory (PEDI), at baseline (T0), 14 (T1) and 28 weeks (T2). RESULTS: Developmental change between T0 and T1 was similar in both groups; change scores in group A and B: GMFM-66 2.4 (SD±2.1) and 3.3 (SD±2.9) (p=0.412); PEDI mobility 8.4 (SD±6.6) and 3.5 (SD±9.2) (p=0.148), respectively. In two children muscle tone increased post-sWBV. 24 children received between 67 and 140 sWBV sessions, rate of completed sessions ranged from 48 to 100% and no dropouts were observed. CONCLUSION: A 14-week home-based sWBV intervention was feasible and safe in toddlers with CP, but was not associated with improvement in gross motor function.


Asunto(s)
Parálisis Cerebral/rehabilitación , Modalidades de Fisioterapia , Vibración/uso terapéutico , Preescolar , Femenino , Humanos , Lactante , Masculino , Modalidades de Fisioterapia/instrumentación , Proyectos Piloto , Vibración/efectos adversos
3.
J Musculoskelet Neuronal Interact ; 16(1): 24-32, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26944820

RESUMEN

OBJECTIVES: Osteogenesis imperfecta (OI) is a rare hereditary disease leading to bone fragility. Denosumab as a RANK ligand antibody inhibiting osteoclast maturation has been approved for osteoporosis treatment in adults. Aim of this study was a 48-week, open-label, pilot study of the safety and efficacy of denosumab in 10 children with OI. METHODS: Ten patients (age range: 5.0-11.0 years; at least two years of prior bisphosphonate treatment) with genetically confirmed OI were studied. Denosumab was administered subcutaneously every 12 weeks with 1 mg/kg body weight. Primary endpoint was change of areal bone mineral density (aBMD) using dual energy x-ray absorptiometry of the lumbar spine after 48 weeks. Safety was assessed by bone metabolism markers and adverse event reporting. RESULTS: Mean relative change of lumbar aBMD was +19 % (95%-CI: 7-31%). Lumbar spine aBMD Z-Scores increased from -2.23±2.03 (mean±SD) to -1.27±2.37 (p=0.0006). Mobility did not change (GMFM-88 +2.72±4.62% (p=0.16); one-minute walking test +11.00±15.82 m (p=0.15). No severe side effects occurred. CONCLUSIONS: On average, there was a significant increase in lumbar spine aBMD percent change after 48 weeks of denosumab. There was no change in mobility parameters and no serious adverse events. Further trials are necessary to assess long-term side effects and efficacy.


Asunto(s)
Conservadores de la Densidad Ósea/uso terapéutico , Denosumab/uso terapéutico , Osteogénesis Imperfecta/tratamiento farmacológico , Absorciometría de Fotón , Densidad Ósea/efectos de los fármacos , Niño , Preescolar , Femenino , Humanos , Vértebras Lumbares/efectos de los fármacos , Masculino , Proyectos Piloto
5.
Cell Death Dis ; 4: e586, 2013 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-23579273

RESUMEN

Neuroblastoma is an embryonal malignancy of the sympathetic nervous system. Spontaneous regression and differentiation of neuroblastoma is observed in a subset of patients, and has been suggested to represent delayed activation of physiologic molecular programs of fetal neuroblasts. Homeobox genes constitute an important family of transcription factors, which play a fundamental role in morphogenesis and cell differentiation during embryogenesis. In this study, we demonstrate that expression of the majority of the human HOX class I homeobox genes is significantly associated with clinical covariates in neuroblastoma using microarray expression data of 649 primary tumors. Moreover, a HOX gene expression-based classifier predicted neuroblastoma patient outcome independently of age, stage and MYCN amplification status. Among all HOX genes, HOXC9 expression was most prominently associated with favorable prognostic markers. Most notably, elevated HOXC9 expression was significantly associated with spontaneous regression in infant neuroblastoma. Re-expression of HOXC9 in three neuroblastoma cell lines led to a significant reduction in cell viability, and abrogated tumor growth almost completely in neuroblastoma xenografts. Neuroblastoma growth arrest was related to the induction of programmed cell death, as indicated by an increase in the sub-G1 fraction and translocation of phosphatidylserine to the outer membrane. Programmed cell death was associated with the release of cytochrome c from the mitochondria into the cytosol and activation of the intrinsic cascade of caspases, indicating that HOXC9 re-expression triggers the intrinsic apoptotic pathway. Collectively, our results show a strong prognostic impact of HOX gene expression in neuroblastoma, and may point towards a role of Hox-C9 in neuroblastoma spontaneous regression.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/genética , Regresión Neoplásica Espontánea/genética , Neoplasias del Sistema Nervioso/genética , Neuroblastoma/genética , Apoptosis/genética , Caspasas/genética , Caspasas/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Preescolar , Citocromos c/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Lactante , Mitocondrias/metabolismo , Mitocondrias/patología , Proteína Proto-Oncogénica N-Myc , Estadificación de Neoplasias , Neoplasias del Sistema Nervioso/metabolismo , Neoplasias del Sistema Nervioso/mortalidad , Neoplasias del Sistema Nervioso/patología , Neuroblastoma/metabolismo , Neuroblastoma/mortalidad , Neuroblastoma/patología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Pronóstico , Transducción de Señal , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Oncogene ; 29(6): 865-75, 2010 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-19901960

RESUMEN

Imbalances in chromosome 11q occur in approximately 30% of primary neuroblastoma and are associated with poor outcome. It has been suggested that 11q loss constitutes a distinct clinico-genetic neuroblastoma subgroup by affecting expression levels of corresponding genes. This study analysed the relationship of 11q loss, clinical phenotype and global transcriptomic profiles in four clinico-genetic subgroups (11q alteration/favourable outcome, n=7; 11q alteration/unfavourable outcome, n=14; no 11q alteration/favourable outcome, n=81; no 11q alteration/unfavourable outcome, n=8; tumours with MYCN amplification and/or 1p loss were excluded). Unsupervised and supervised comparisons of gene expression profiles consistently showed significantly different mRNA patterns between favourable and unfavourable neuroblastomas, both in the subgroups with and without 11q loss. In contrast, favourable tumours with and without 11q loss showed highly similar transcriptomic profiles. Disproportionate downregulation of 11q genes was observed only in unfavourable tumours with 11q loss. The diverging molecular profiles were neither caused by considerable differences in the size of the deleted regions nor by differential methylation patterns of 11q genes. Together, this study shows that neuroblastoma with 11q loss comprises two biological subgroups that differ both in their clinical phenotype and gene expression patterns, indicating that 11q loss is not a primary determinant of neuroblastoma tumour behaviour.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 11/genética , Biología Computacional , Perfilación de la Expresión Génica , Genómica , Neuroblastoma/diagnóstico , Neuroblastoma/genética , Preescolar , Cromosomas Humanos Par 11/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Metilación , Neuroblastoma/metabolismo , Neuroblastoma/patología , Pronóstico , Regiones Promotoras Genéticas/genética
7.
Nuklearmedizin ; 47(3): 132-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18493694

RESUMEN

The German translation of the EANM guideline for MIBG scintigraphy in children (Olivier P et al. EJNM MI 2003; 30: B45-B50; Hahn K. Der Nuklearmediziner 2002; 25: 101-105) was reviewed and actualized according to current publications, legal requirements and conditions in Germany. For the first time this guideline was generated in consensus with the neuroblastoma study group of the Association of Paediatric Haematologie and Oncology (GPOH) with the result of an interdisciplinary recommendation. Further main alterations are related to the recommended (123)I activities with respect to the new EANM Paediatric Dosage Card and the explicit recommendation of SPECT.


Asunto(s)
3-Yodobencilguanidina , Neoplasias de las Glándulas Suprarrenales/diagnóstico por imagen , Neuroblastoma/diagnóstico por imagen , Feocromocitoma/diagnóstico por imagen , Tomografía Computarizada de Emisión de Fotón Único/métodos , Niño , Neoplasias Hematológicas/diagnóstico por imagen , Humanos , Guías de Práctica Clínica como Asunto , Tomografía Computarizada de Emisión de Fotón Único/normas
8.
Oncogene ; 27(23): 3329-38, 2008 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-18084322

RESUMEN

Cell adhesion molecule 1 (CADM1) is a putative tumour suppressor gene, which is downregulated in many solid tumours. In neuroblastoma, loss of CADM1 expression has recently been found in disseminated tumours with adverse outcome, prompting us to investigate its role in neuroblastoma tumour progression. Oligonucleotide-microarray analysis of 251 neuroblastoma specimens demonstrated that CADM1 downregulation is associated with unfavourable prognostic markers like disseminated stage 4, age >18 months, MYCN amplification and chromosome 11q alterations (P<0.001 each). Furthermore, low CADM1 expression was significantly correlated with unfavourable gene expression-based classification (P<0.001) and adverse patient outcome (P<0.001). Bisulphite sequencing and genetic analysis of 18 primary neuroblastomas suggested that neither haploinsufficiency nor hypermethylation is regularly involved in CADM1 gene silencing in neuroblastoma, which is in contrast to results obtained in other malignancies. In addition, no mutations disrupting the CADM1 reading frame were found in 25 primary neuroblastomas. Over-expression of CADM1 in neuroblastoma cells resulted in significant reduction of proliferation, viability and colony formation in soft agar. Collectively, our results suggest that downregulation of CADM1 tumour suppressor gene expression is a critical event in neuroblastoma pathogenesis resulting in tumour progression and unfavourable patient outcome.


Asunto(s)
Inmunoglobulinas/genética , Proteínas de la Membrana/genética , Neuroblastoma/diagnóstico , Neuroblastoma/genética , Proteínas Supresoras de Tumor/genética , Adolescente , Adulto , Biomarcadores de Tumor/genética , Molécula 1 de Adhesión Celular , Moléculas de Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/genética , Niño , Preescolar , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor/fisiología , Humanos , Inmunoglobulinas/fisiología , Lactante , Recién Nacido , Proteínas de la Membrana/fisiología , Mutación/fisiología , Neuroblastoma/mortalidad , Neuroblastoma/patología , Pronóstico , Regiones Promotoras Genéticas , ARN Mensajero/análisis , Análisis de Supervivencia , Proteínas Supresoras de Tumor/fisiología
9.
Eur J Paediatr Neurol ; 12(1): 51-5, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17625938

RESUMEN

Opsoclonus-myoclonus syndrome (OMS) is a rare and debilitating disorder of unknown etiology affecting children and adults. Outcome is unfavourable; approximately 80% of children with OMS suffer from mild to severe neurological handicaps, mainly cognitive impairment. A standard therapy does not exist. Due to the possible immune-mediated mechanisms, treatment with steroids, ACTH, plasmapheresis and immunoglobulins can be successful. However, some children become steroid dependent and symptoms may reoccur after treatment has been finished. We present two girls with OMS, who had a prolonged clinical course lasting 4 and 9 years with many relapses. Both children developed symptoms around the age of two years. Diagnostic work-up to exclude neuroblastoma was negative. Several treatment modalities including oral steroids, dexamethasone pulses, immunoglobulin and cyclosporine were used without lasting success. In addition, cognitive impairment developed in both children. In order to prevent further clinical and mental deterioration, 6 pulses of cyclophosphamide in combination with dexamethasone pulses every 4 weeks were administered. Both children showed significant improvement of OMS symptoms. One girl is still symptom free 18 months after treatment, mild ataxia developed in the other after 12 months. Both children are mentally handicapped and in special need schools. We conclude that combination of cyclophosphamide pulses and dexamethasone pulse therapy is a therapeutic option even after a long clinical course to improve symptoms of OMS.


Asunto(s)
Antiinflamatorios/uso terapéutico , Ciclofosfamida/uso terapéutico , Dexametasona/uso terapéutico , Inmunosupresores/uso terapéutico , Síndrome de Opsoclonía-Mioclonía/tratamiento farmacológico , Adolescente , Preescolar , Quimioterapia Combinada , Femenino , Escritura Manual , Humanos , Imagen por Resonancia Magnética , Destreza Motora/efectos de los fármacos , Síndrome de Opsoclonía-Mioclonía/patología , Prevención Secundaria
10.
Neuropediatrics ; 38(3): 114-6, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17985258

RESUMEN

Opsoclonus-myoclonus syndrome (OMS) is a rare neurological disease in childhood which can be associated with neuroblastoma. Since autoantibodies have been detected in some patients with OMS, an autoimmune etiology is suspected. We compared the prevalence of autoimmune disorders and autoantibodies in parents of children with OMS and in a group of controls of same age and sex. Autoimmune diseases were found in 15.8% of the parents of OMS children, but only in 2.0% of the controls (p<0.001) There was also an increased prevalence of autoantibodies in the OMS parents (42.8% vs. 8.0%, p<0.001). Thyroid diseases were the most frequent autoimmune diseases found, followed by inflammatory rheumatic diseases. Interestingly, the OMS parents also had significantly more autoantibodies against CNS structures than the controls (p<0.01). These findings support the autoimmune hypothesis of childhood OMS and may also hint to a genetic susceptibility for OMS.


Asunto(s)
Autoanticuerpos/inmunología , Enfermedades Autoinmunes/epidemiología , Enfermedades Autoinmunes/inmunología , Síndrome de Opsoclonía-Mioclonía/epidemiología , Síndrome de Opsoclonía-Mioclonía/inmunología , Padres , Adulto , Anticuerpos Antinucleares/sangre , Estudios de Casos y Controles , Niño , Femenino , Humanos , Inmunoglobulinas Estimulantes de la Tiroides/sangre , Masculino , Neuroblastoma/inmunología , Prevalencia
11.
Nuklearmedizin ; 45(6): 254-61, 2006.
Artículo en Alemán | MEDLINE | ID: mdl-17149494

RESUMEN

AIM: Targeted radiotherapies using iodine-131 meta-iodobenzylguanidin have long been in use for treatment of stage IV neuroblastoma but reliable dosimetric data are scarce. METHOD: This work presents an approach to determine the whole body exposure and tumour doses delivered during therapy. Dosimetric data are reported and discussed for six treatments carried out according to the trial protocol NB2004 as it is in use in our study in the last two years. RESULTS: Whole body exposures are found to be in the range of 1.75 to 2.5 Gy whereas tumour doses vary between 15 and 55 Gy. CONCLUSION: The course of action prescribed by the trial protocol allows whole body exposure as well as tumour doses to be determined routinely.


Asunto(s)
3-Yodobencilguanidina/uso terapéutico , Neuroblastoma/radioterapia , 3-Yodobencilguanidina/farmacocinética , Humanos , Radioisótopos de Yodo/farmacocinética , Radioisótopos de Yodo/uso terapéutico , Tasa de Depuración Metabólica , Distribución Tisular
12.
Nuklearmedizin ; 45(4): 145-51; quiz N39-40, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16964339

RESUMEN

AIM: (131)I-meta-iodobenzylguanidine ((131)I-MIBG) therapy has been used in neuroblastoma treatment for many years but its value in high intensive first line treatment protocols is not exactly known. PATIENTS, METHODS: Stage 4 neuroblastoma patients >1 year with (123)I-MIBG positive residual disease (primary tumour and/or metastasis) after complete induction chemotherapy according to the German neuroblastoma trial NB97 were retrospectively analyzed. RESULTS: One-hundred-eleven patients had (123)I-MIBG positive residual disease after complete induction chemotherapy. Forty patients received (131)I-MIBG therapy using a median activity of 0.44 GBq/kg body weight. By univariate analysis, patients who underwent (131)I-MIBG therapy had a better 3-year event free survival (3-y-EFS 46 +/- 8%) and 3-year overall survival (3-y-OS 58 +/- 9%) than 71 patients without (131)I-MIBG therapy (3-y-EFS 19 +/- 5%, p = 0.003; 3-y-OS 43 +/- 6%, p = 0.037). However, subgroup analysis of 66 patients who underwent high dose chemotherapy with autologous stem cell transplantation (ASCT) during treatment found a very similar outcome with (131)I-MIBG therapy (3-y-EFS 49 +/- 9%, 3-y-OS 59 +/- 10%) and without (131)I-MIBG therapy (3-y-EFS 33 +/- 9%, p = 0.171; 3-y-OS 59 +/- 9%, p = 0.285) due to the dominating effect of ASCT. By multivariate analysis, (131)I-MIBG therapy had no impact on EFS (p = 0.494) and OS (p = 0.891). Only ASCT, external beam radiation therapy and MYCN amplification were important for EFS and OS. CONCLUSIONS: An independent advantage of I-131-MIBG therapy could not be proven in this retrospective analysis. The ongoing German Neuroblastoma Trial NB2004 will address the influence of (131)I-MIBG therapy with emphasis on tumour dosimetry.


Asunto(s)
3-Yodobencilguanidina/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia , Radioisótopos de Yodo/uso terapéutico , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/radioterapia , Adolescente , Adulto , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Niño , Preescolar , Supervivencia sin Enfermedad , Estudios de Seguimiento , Humanos , Estadificación de Neoplasias , Neuroblastoma/mortalidad , Neuroblastoma/patología , Análisis de Regresión , Estudios Retrospectivos , Análisis de Supervivencia , Resultado del Tratamiento
13.
Klin Padiatr ; 218(3): 132-8, 2006.
Artículo en Alemán | MEDLINE | ID: mdl-16688668

RESUMEN

Availability of statistically sufficient numbers of tumor samples and other biomaterials in high quality together with corresponding clinical data is crucial for biomedical research. Tumor repositories from individual scientists are mostly not sufficient to satisfy these criteria, especially since pediatric tumors are rare. In 2000 three centralized tumor repositories (neuroblastoma in Cologne, nephroblastoma in Würzburg, hepatoblastoma, brain tumors in Bonn) have been established by the "German Competence Net Pediatric Oncology und Hematology". The aim was to collect biomaterial including tumor samples, normal tissue, and blood in high quality for research and diagnostic purposes at a central institution. Informed consent of the parents or patients is a prerequisite for scientific use of the samples and is requested by the therapy trial. The samples are collected according to accepted standards and shipped in the specially designed Tumorbox. The tumor repository organizes the distribution of the samples to the cooperating diagnostic laboratories. The number of collected tumor samples has increased over the years. In 2000, samples from 200 patients were collected while the patient number increased to 321 in 2005. Over the years the tumor repositories collected more than 7,150 samples (fresh frozen tumor, fresh frozen normal tissue, and blood). Through links with clinical trial databases the samples can be connected with clinical data. 12 of 14 applications for tumor material to be used in specific scientific projects have been approved by an independent supervisory board. The establishment of central tumor repositories represents a major step for biomedical research activities and quality control in pediatric oncology.


Asunto(s)
Materiales Biocompatibles/provisión & distribución , Investigación Biomédica/estadística & datos numéricos , Bases de Datos Genéticas/provisión & distribución , Necesidades y Demandas de Servicios de Salud/estadística & datos numéricos , Neoplasias de Células Germinales y Embrionarias/patología , Bancos de Tejidos/provisión & distribución , Neoplasias Encefálicas/epidemiología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Niño , Ensayos Clínicos como Asunto/tendencias , Predicción , Secciones por Congelación , Alemania , Hepatoblastoma/epidemiología , Hepatoblastoma/genética , Hepatoblastoma/patología , Humanos , Neoplasias Renales/epidemiología , Neoplasias Renales/genética , Neoplasias Renales/patología , Neoplasias Hepáticas/epidemiología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias de Células Germinales y Embrionarias/epidemiología , Neoplasias de Células Germinales y Embrionarias/genética , Neuroblastoma/epidemiología , Neuroblastoma/genética , Neuroblastoma/patología , Tumor de Wilms/epidemiología , Tumor de Wilms/genética , Tumor de Wilms/patología
14.
Neuropediatrics ; 37(5): 291-5, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17236108

RESUMEN

Opsoclonus-myoclonus syndrome (OMS) is a rare movement disorder characterized by chaotic eye movements, myoclonus, and ataxia associated with severe irritability. Different treatment modalities including steroids and cyclophosphamide have been tried in the past often with significant side effects and variable success. Here we present 11 children, diagnosed with OMS between 1999 and 2005 and treated with high dose dexamethasone pulses. Main symptoms at presentation were opsoclonus (11/11), ataxia and/or myoclonus (11/11), irritability (10/11) associated with a neuroblastoma in four children. Number of dexamethasone pulses ranged from 6 to 60 pulses. No major side effects were reported. In 6/11 children a complete and sustained remission of OMS symptoms was achieved after 6 to 29 pulses of dexamethasone. Two children from this group have a normal development and no neurological sequelae. Two further children have minor delays in fine- and gross-motor skills. Two children despite a complete recovery of OMS symptoms have persisting developmental problems. 5/11 children still require regular dexamethasone pulses in addition to daily prednisolone (n = 1) or have received cyclophosphamide pulses meanwhile (n = 2). All children continue to have developmental and neurological difficulties. In summary treatment with high dose pulsatile dexamethasone appears to be safe and beneficial in a subgroup of patients with OMS.


Asunto(s)
Dexametasona/uso terapéutico , Glucocorticoides/uso terapéutico , Síndrome de Opsoclonía-Mioclonía/tratamiento farmacológico , Preescolar , Vías de Administración de Medicamentos , Femenino , Humanos , Lactante , Masculino , Pruebas Neuropsicológicas , Estudios Retrospectivos
15.
Klin Padiatr ; 217(3): 147-52, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15858706

RESUMEN

BACKGROUND: Antibody treatment is considered tolerable and potentially effective in the therapy of neuroblastoma. We have analyzed the clinical data of infants < 1 year with stage 4 neuroblastoma with regard to the consolidation treatment. PATIENTS AND METHODS: Infants < 1 year with stage 4 neuroblastoma who completed initial treatment (6-8 chemotherapy cycles followed either by 4 cycles low dose oral chemotherapy or high dose chemotherapy with stem cell transplantation) without event were eligible for this trial. Consolidation therapy consisted of 6 cycles of antibody ch14.18 (20 mg/m(2) x d ch14.18 for 5 days every 2 months) or 12 months oral maintenance chemotherapy (MT). RESULTS: Of 59 evaluable patients, 31 received a total of 159 ch14.18 cycles, 16 received MT instead, and 12 had no further treatment. Fever (47 % of cycles), abnormal CRP without infection (25 %), rash (23 %), cough (16 %), and pain (8 %) were the main side effects. Univariate analysis found no difference in event free survival (3-year-EFS 80.5 +/- 7.1 %, 87.5 +/- 8.3 %, and 75.0 +/- 12.5 % for patients treated with antibody ch14.18, MT, and no further therapy, p = 0.433) and overall survival (3-year-OS 90.1 +/- 5.4 %, 93.8 +/- 6.0 %, and 91.7 +/- 8.0 % for patients treated with antibody ch14.18, MT, and no further therapy, p = 0.931). Multivariate analysis failed to demonstrate an advantage of antibody treatment. CONCLUSION: The outcome of infants with stage 4 neuroblastoma is good. Consolidation treatment with ch14.18 was tolerable but associated with fever, elevated CRP, rash, cough, and pain as side effects. Compared to oral maintenance chemotherapy and no consolidation treatment, ch14.18 treatment had no impact on the patients' outcome which confirms the results found in children > 1 year.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Inmunoterapia , Neuroblastoma/terapia , Administración Oral , Factores de Edad , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/efectos adversos , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Terapia Combinada , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Análisis Multivariante , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/mortalidad , Estudios Retrospectivos , Trasplante de Células Madre , Análisis de Supervivencia , Resultado del Tratamiento , Tretinoina/administración & dosificación , Tretinoina/uso terapéutico
16.
Eur J Cancer ; 40(18): 2753-9, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15571958

RESUMEN

While the role of MYCN-amplification (MNA) for risk assessment in neuroblastoma is undisputed, the phenomenon of gene copy excess below the amplification threshold is rarely described. To discuss biological characteristics and the clinical impact of the so-called MYCN-gain versus amplified or non-amplified cases, we investigated the MYCN status of 659 patients uniformly analysed by fluorescence in situ hybridisation. The number of MYCN-amplified tumours in our cohort was 18% (116/659); an additional 38 tumours (6%) displayed MYCN-gain. Both alterations were associated with an advanced stage disease, an increased patient age and further chromosomal alterations. Most of the amplified neuroblastomas displayed 1p aberrations, whereas MYCN-gain tumours correlated with 11q alterations. In contrast to the amplified cases, tumours with gain displayed no increased MYCN RNA levels. MNA versus non-amplification discriminated between good and poor outcomes, independent of stage, age and the degree of amplification. However, patients with amplified tumours showed a significantly better outcome when this was combined with non-stage 4 disease and age <1 year versus stage 4 and age < 1 year. Although MYCN-gain was associated with poor event-free-survival (EFS) in stages 1-3, 4S (P=0.005), this might be related to associated genetic aberrations and not to the MYCN-gain itself. A survival difference between neuroblastomas with gain and single copy MYCN could not be delineated. In conclusion, MNA predicts a poor outcome for neuroblastoma patients of all stages and age. MYCN-gain is also a characteristic feature of advanced stage tumours and older patients, but is not associated with higher MYCN expression and appears not to be discriminative in predicting patient outcome.


Asunto(s)
Amplificación de Genes/genética , Genes myc , Neuroblastoma/genética , Southern Blotting , Preescolar , Supervivencia sin Enfermedad , Humanos , Lactante , Recurrencia Local de Neoplasia/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Estadísticas no Paramétricas
17.
Eur J Cancer ; 39(13): 1899-903, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12932669

RESUMEN

The value of the tumour markers vanillylmandelic acid (VMA) and homovanillic acid (HVA) in urine (u) and serum (s), neurone-specific enolase (NSE), and lactate dehydrogenase (LDH) in the early prediction of relapse/progression in neuroblastoma is not known. We analysed the data of neuroblastoma patients who had successfully completed first-line treatment and had laboratory results available from their initial diagnosis and from relapse/progression (n=196). Patients' overall survival from relapse or progression was 21.5+/-4.2% (mean+/-standard deviation). At diagnosis, we found abnormal results in 75% for VMA and/or HVA (s), 92% for VMA and/or HVA (u), 90% for NSE, and 81% for LDH. We found a lower incidence of abnormal results at relapse or progression with 40% for VMA and/or HVA (s), 54% for HVA and/or VMA (u), 61% for NSE, and 48% for LDH. Sensitivity of all markers was higher for metastatic compared with local recurrence. NSE was the best, being able to detect 42% of the localised relapses, 77% of the combined local/metastatic relapses, and 69% of the metastatic recurrences. Relapse or progression in neuroblastoma cannot be detected reliably by monitoring tumour markers alone. Therefore, follow-up of neuroblastoma patients must include clinical assessment and imaging studies.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias de la Médula Ósea/secundario , Recurrencia Local de Neoplasia/diagnóstico , Neuroblastoma/diagnóstico , Adolescente , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/orina , Neoplasias de la Médula Ósea/sangre , Neoplasias de la Médula Ósea/orina , Niño , Preescolar , Ensayos Clínicos como Asunto , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Estudios de Seguimiento , Ácido Homovanílico/sangre , Ácido Homovanílico/orina , Humanos , Lactante , Recién Nacido , Radioisótopos de Yodo , Yodobencenos , L-Lactato Deshidrogenasa/sangre , L-Lactato Deshidrogenasa/orina , Imagen por Resonancia Magnética , Recurrencia Local de Neoplasia/mortalidad , Neuroblastoma/mortalidad , Fosfopiruvato Hidratasa/sangre , Fosfopiruvato Hidratasa/orina , Estudios Retrospectivos , Sensibilidad y Especificidad , Tomografía Computarizada por Rayos X , Ácido Vanilmandélico/sangre , Ácido Vanilmandélico/orina
18.
Cancer Lett ; 197(1-2): 11-7, 2003 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-12880954

RESUMEN

During the last two decades new diagnostic and therapeutic tools have been utilized to improve the poor survival chances of children with stage 4 neuroblastoma. This study reviews the risk profiles and the long-term outcome of patients from five consecutive German neuroblastoma trials. A total of 96% of all German patients registered at the German childhood cancer registry with neuroblastoma stage 4 over 1 year of age at diagnosis entered one of the trials during 1979-2001. Eight hundred and twenty-eight consecutive children were analyzed retrospectively. In spite of having significantly improved diagnostic tools like bone marrow superstaging and mIBG scintigraphy the stage 4 incidence did not increase after reaching completeness of the registry (5.4 cases/100,000 children at 1-14 years of age; P=0.52). The distribution of the primary tumors and of metastases was constant over the periods. The amount of bone marrow infiltration did not change with time. The risk factors lactate dehydrogenase, ferritin and MYCN, and the clinical risk groups 4A, 4B, 4C also remained constant over the trials with a few exceptions for NB97. The 5-year event free survival increased from 0.01+/-0.01 (NB79) to 0.14+/-0.03 (NB85), 0.16+/-0.04 (NB82), 0.27+/-0.02 (NB90), and 0.33+/-0.04 (NB97). The overall survival rates improved similarly from 0.04 (NB79) to 0.44 (NB97). In conclusion, the improved survival was associated with better treatment and not caused by lower risk profiles in stage 4 neuroblastoma patients.


Asunto(s)
Neuroblastoma/diagnóstico , Neuroblastoma/epidemiología , Adolescente , Distribución por Edad , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Niño , Preescolar , Ensayos Clínicos como Asunto , Ferritinas/metabolismo , Genes myc/fisiología , Alemania/epidemiología , Humanos , Lactante , Recién Nacido , L-Lactato Deshidrogenasa/metabolismo , Estadificación de Neoplasias , Neuroblastoma/tratamiento farmacológico , Pronóstico , Estudios Retrospectivos , Factores de Riesgo , Tasa de Supervivencia , Resultado del Tratamiento
19.
Bone Marrow Transplant ; 30(12): 893-8, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12476282

RESUMEN

Children with Wilms tumor who have a particular risk of failure at relapse or at primary diagnosis were treated with high-dose chemotherapy (HDC) and autologous peripheral blood stem cell rescue in order to improve their probability of survival. From April 1992 to December 1998, 23 evaluable patients received HDC within the German Cooperative Wilms Tumor Studies. Nineteen were given melphalan, etoposide and carboplatin (MEC); the others received different regimens. The dose of carboplatin was adjusted according to renal function. Indications for HDC were high-risk relapse in 20 patients, bone metastases in two patients and no response in one patient. Fourteen of 23 patients are alive after a median observation time of 41 months, 11 of 14 in continuous complete remission, three in CR after relapse post HDC. The estimated survival and event-free survival for these patients are 60.9% and 48.2%. Twelve children relapsed after HDC; nine of them died within 12 months and three are surviving from 20 to 33 months after relapse. The main toxicities were hematologic, mucositis and renal (tubular dysfunction; intermittent hemodialysis in one patient). There were no toxic deaths. About half of the children suffering from Wilms tumor with very unfavorable prognostic factors survive disease-free after HDC for over 3 years. Besides hematological toxicity, mucositis and infections, renal function is at risk during HDC. With dose adjustment on glomerular filtration rate, however, no permanent renal failure was observed.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ciclofosfamida/análogos & derivados , Neoplasias Renales/tratamiento farmacológico , Trasplante de Células Madre de Sangre Periférica , Tumor de Wilms/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carboplatino/administración & dosificación , Carboplatino/efectos adversos , Niño , Preescolar , Terapia Combinada , Ciclofosfamida/administración & dosificación , Supervivencia sin Enfermedad , Etopósido/administración & dosificación , Etopósido/efectos adversos , Femenino , Alemania/epidemiología , Enfermedades Hematológicas/inducido químicamente , Humanos , Ifosfamida/administración & dosificación , Lactante , Enfermedades Renales/inducido químicamente , Neoplasias Renales/mortalidad , Neoplasias Renales/terapia , Tablas de Vida , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Masculino , Melfalán/administración & dosificación , Melfalán/efectos adversos , Pronóstico , Estomatitis/inducido químicamente , Análisis de Supervivencia , Tiotepa/administración & dosificación , Trasplante Autólogo , Tumor de Wilms/mortalidad , Tumor de Wilms/secundario , Tumor de Wilms/terapia
20.
Klin Padiatr ; 214(4): 149-52, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12165893

RESUMEN

BACKGROUND: Platinum compound based chemotherapy has contributed to improved survival rates in neuroblastoma patients. We studied the association of hearing loss with the use of platinum based drugs. METHODS: Data of children from the two German neuroblastoma study cohorts, NB90 and NB97, were analyzed for the incidence of hearing impairments greater than WHO grade 2. Data from patients surviving more than 1 year after diagnosis without progression or recurrence were used. RESULTS: Of these 1,170 patients, 146 (12.5 %) had persisting hearing impairments. The incidence was low after localized neuroblastoma INSS stage 1 - 3 with 35/650 (5 %) and stage 4S with 2/113 (2 %), but high in stage 4 disease with 109/405 (27 %). Ototoxicity depended on the cumulative cisplatin dose. For doses of 1 - 200 mg/m (2), 201 - 400 mg/m (2), 401 - 600 mg/m (2) and 601 - 800 mg/m (2) we found hearing impairments in 5/39 (12 %), 28/221 (13 %), 61/230 (26 %) and 50/225 (22 %) patients, respectively. The incidence of hearing impairment was not age dependent. In stage 4 patients, there was an additional effect of high dosed carboplatin. 65/188 (40 %) patients developed hearing impairments after carboplatin containing (1,500 mg/m (2)) high dose chemotherapy with autologous stem cell reinfusion compared to only 33/217 (15 %) in patients receiving platinum free maintenance chemotherapy. Substitution of cisplatin (40 mg/m (2) x d, d 1 - 4, 96 h infusion) with carboplatin (100 mg/m (2) x d, d 1 - 4, 1 - 2 h infusion) due to otoxicity during induction chemotherapy did not reduce event free survival. CONCLUSION: One fourth of high risk neuroblastoma survivors suffer from treatment induced hearing impairments. The substitution of cisplatin with carboplatin was not associated with an increased rate of tumor recurrences.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carboplatino/efectos adversos , Cisplatino/efectos adversos , Pérdida Auditiva Sensorineural/inducido químicamente , Neuroblastoma/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carboplatino/administración & dosificación , Niño , Preescolar , Cisplatino/administración & dosificación , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Estudios de Seguimiento , Pérdida Auditiva Sensorineural/diagnóstico , Humanos , Lactante , Masculino , Estadificación de Neoplasias , Neuroblastoma/patología , Pronóstico , Factores de Riesgo , Trasplante de Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...