Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cancer ; 130(3): 467-475, 2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-37788149

RESUMEN

BACKGROUND: Overgrowth syndromes (e.g., Beckwith-Wiedemann) are associated with an increased risk of pediatric cancer, although there are few population-based estimates of risk. There are also limited studies describing associations between other overgrowth features (e.g., hepatosplenomegaly) and pediatric cancer. Therefore, cancer risk among children with these conditions was evaluated with data from a large, diverse population-based registry linkage study. METHODS: This study includes all live births in Texas during the years 1999-2017. Children with overgrowth features and syndromes were identified from the Texas Birth Defects Registry; children with cancer were identified by linkage to the Texas Cancer Registry. Cox regression models were used to estimate the hazard ratio (HR) and 95% confidence interval (CI) for the association between each overgrowth syndrome/feature and cancer, which were adjusted for infant sex and maternal age. RESULTS: In the total birth cohort (n = 6,997,422), 21,207 children were identified as having an overgrowth syndrome or feature. Children with Beckwith-Wiedemann syndrome were 42 times more likely to develop pediatric cancer (95% CI, 24.20-71.83), with hepatoblastoma being the most common, followed by Wilms tumor. The presence of any isolated overgrowth feature was associated with increased cancer risk (HR, 4.70; 95% CI, 3.83-5.77); associations were strongest for hepatosplenomegaly (HR, 23.04; 95% CI, 13.37-39.69) and macroglossia (HR, 11.18; 95% CI, 6.35-19.70). CONCLUSIONS: This population-based assessment confirmed prior findings that children with either overgrowth syndromes or features were significantly more likely to develop cancer. Overall, this study supports recommendations for cancer surveillance in children with these conditions and may also inform future research into cancer etiology.


Asunto(s)
Síndrome de Beckwith-Wiedemann , Neoplasias Renales , Neoplasias Hepáticas , Tumor de Wilms , Lactante , Niño , Humanos , Incidencia , Síndrome de Beckwith-Wiedemann/complicaciones , Síndrome de Beckwith-Wiedemann/epidemiología , Síndrome de Beckwith-Wiedemann/genética , Tumor de Wilms/epidemiología , Neoplasias Renales/complicaciones , Neoplasias Hepáticas/complicaciones
2.
Pediatr Blood Cancer ; 68(10): e29178, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34133064

RESUMEN

BACKGROUND: Patients with chiasmatic-hypothalamic low-grade glioma (CHLGG) have frequent MRIs with gadolinium-based contrast agents (GBCA) for disease monitoring. Cumulative gadolinium deposition in the brains of children is a potential concern. The purpose of this study is to evaluate whether MRI with GBCA is necessary for determining radiographic tumor progression in children with CHLGG. METHODS: Children who were treated for progressive CHLGG from 2005 to 2019 at Texas Children's Cancer Center were identified. Pre- and post-contrast MRI sequences were separately reviewed by one neuroradiologist who was blinded to the clinical course. Three dimensional measurements and tumor characteristics were evaluated. Radiographic progression was defined as a 25% increase in size (product of two largest dimensions) compared with baseline or best response after initiation of therapy. RESULTS: A total of 28 patients with progressive CHLGG were identified with a total of 683 MRIs with GBCA reviewed (mean 24 MRIs/patient; range, 11-43 MRIs). Radiographic progression was observed 92 times, 91 (99%) on noncontrast and 90 (98%) on contrast imaging. Sixty-seven progressions necessitating management changes were identified in all (100%) noncontrast sequences and 66 (99%) contrast sequences. Tumor growth > 2 mm in any dimension was identified in 184/187 (98%) noncontrast and 181/187 (97%) with contrast imaging. Metastatic tumors were better visualized on contrast imaging in 4/7 (57%). CONCLUSION: MRI without GBCA effectively identifies patients with progressive disease. When imaging children with CHLGG, eliminating GBCA should be considered unless monitoring patients with metastatic disease.


Asunto(s)
Gadolinio , Glioma , Encéfalo/diagnóstico por imagen , Niño , Medios de Contraste , Glioma/diagnóstico por imagen , Humanos , Imagen por Resonancia Magnética , Estudios Retrospectivos
3.
Am J Hum Genet ; 101(4): 503-515, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28942966

RESUMEN

Bromodomain PHD finger transcription factor (BPTF) is the largest subunit of nucleosome remodeling factor (NURF), a member of the ISWI chromatin-remodeling complex. However, the clinical consequences of disruption of this complex remain largely uncharacterized. BPTF is required for anterior-posterior axis formation of the mouse embryo and was shown to promote posterior neuroectodermal fate by enhancing Smad2-activated wnt8 expression in zebrafish. Here, we report eight loss-of-function and two missense variants (eight de novo and two of unknown origin) in BPTF on 17q24.2. The BPTF variants were found in unrelated individuals aged between 2.1 and 13 years, who manifest variable degrees of developmental delay/intellectual disability (10/10), speech delay (10/10), postnatal microcephaly (7/9), and dysmorphic features (9/10). Using CRISPR-Cas9 genome editing of bptf in zebrafish to induce a loss of gene function, we observed a significant reduction in head size of F0 mutants compared to control larvae. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and phospho-histone H3 (PH3) staining to assess apoptosis and cell proliferation, respectively, showed a significant increase in cell death in F0 mutants compared to controls. Additionally, we observed a substantial increase of the ceratohyal angle of the craniofacial skeleton in bptf F0 mutants, indicating abnormal craniofacial patterning. Taken together, our data demonstrate the pathogenic role of BPTF haploinsufficiency in syndromic neurodevelopmental anomalies and extend the clinical spectrum of human disorders caused by ablation of chromatin remodeling complexes.


Asunto(s)
Anomalías Múltiples/genética , Antígenos Nucleares/genética , Anomalías Craneofaciales/genética , Regulación del Desarrollo de la Expresión Génica , Haploinsuficiencia/genética , Trastornos del Desarrollo del Lenguaje/genética , Microcefalia/genética , Proteínas del Tejido Nervioso/genética , Factores de Transcripción/genética , Anomalías Múltiples/patología , Adolescente , Animales , Antígenos Nucleares/metabolismo , Sistemas CRISPR-Cas , Proliferación Celular , Células Cultivadas , Niño , Preescolar , Ensamble y Desensamble de Cromatina , Estudios de Cohortes , Anomalías Craneofaciales/patología , Femenino , Edición Génica , Haploinsuficiencia/fisiología , Humanos , Trastornos del Desarrollo del Lenguaje/patología , Larva/genética , Larva/crecimiento & desarrollo , Masculino , Microcefalia/patología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuronas/patología , Fenotipo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
4.
Pediatr Blood Cancer ; 67(6): e28258, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32196895

RESUMEN

Germline PTEN (phosphatase and tensin homolog) mutations lead to inappropriate cell survival and growth, and a predisposition to multiple cancers. Some patients also have vascular anomalies (VAs), and it is unclear whether these patients have different phenotypes or oncologic risks. We conducted a two-institution retrospective cohort study to better understand the phenotypes of children and young adults with PTEN mutations, and to compare individuals with VA to those without. Almost half of the patients had thyroid tumors and nearly one quarter developed gastrointestinal tumors before 30 years of age. The presence of VA was positively associated with bulky overgrowth but did not appear to modify oncologic risk.


Asunto(s)
Neoplasias Gastrointestinales/patología , Mutación , Neovascularización Patológica/complicaciones , Fosfohidrolasa PTEN/genética , Neoplasias de la Tiroides/patología , Malformaciones Vasculares/complicaciones , Adolescente , Adulto , Niño , Preescolar , Femenino , Estudios de Seguimiento , Neoplasias Gastrointestinales/etiología , Neoplasias Gastrointestinales/genética , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Fenotipo , Pronóstico , Estudios Retrospectivos , Neoplasias de la Tiroides/etiología , Neoplasias de la Tiroides/genética , Adulto Joven
6.
Genet Med ; 21(4): 867-876, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30190611

RESUMEN

PURPOSE: Neurofibromatosis type 1 (NF1) is characterized by a highly variable clinical presentation, but almost all NF1-affected adults present with cutaneous and/or subcutaneous neurofibromas. Exceptions are individuals heterozygous for the NF1 in-frame deletion, c.2970_2972del (p.Met992del), associated with a mild phenotype without any externally visible tumors. METHODS: A total of 135 individuals from 103 unrelated families, all carrying the constitutional NF1 p.Met992del pathogenic variant and clinically assessed using the same standardized phenotypic checklist form, were included in this study. RESULTS: None of the individuals had externally visible plexiform or histopathologically confirmed cutaneous or subcutaneous neurofibromas. We did not identify any complications, such as symptomatic optic pathway gliomas (OPGs) or symptomatic spinal neurofibromas; however, 4.8% of individuals had nonoptic brain tumors, mostly low-grade and asymptomatic, and 38.8% had cognitive impairment/learning disabilities. In an individual with the NF1 constitutional c.2970_2972del and three astrocytomas, we provided proof that all were NF1-associated tumors given loss of heterozygosity at three intragenic NF1 microsatellite markers and c.2970_2972del. CONCLUSION: We demonstrate that individuals with the NF1 p.Met992del pathogenic variant have a mild NF1 phenotype lacking clinically suspected plexiform, cutaneous, or subcutaneous neurofibromas. However, learning difficulties are clearly part of the phenotypic presentation in these individuals and will require specialized care.


Asunto(s)
Discapacidades para el Aprendizaje/genética , Neurofibroma Plexiforme/genética , Neurofibromatosis 1/genética , Neurofibromina 1/genética , Adolescente , Adulto , Niño , Preescolar , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Heterocigoto , Humanos , Lactante , Discapacidades para el Aprendizaje/fisiopatología , Masculino , Mutación Missense/genética , Neurofibroma Plexiforme/fisiopatología , Neurofibromatosis 1/patología , Eliminación de Secuencia , Adulto Joven
7.
Curr Opin Pediatr ; 31(1): 41-47, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30531401

RESUMEN

PURPOSE OF REVIEW: A significant proportion of pediatric cancer occurs in children with hereditary cancer predisposition syndromes. Their survival may be significantly improved and/or late effects diminished through screening for their greatly elevated cancer risks. Here, an overview of new developments in the field of pediatric cancer surveillance is provided. RECENT FINDINGS: Consensus-based screening guidelines have been developed for most syndromes associated with childhood cancer risks. Studies evaluating the clinical utility of these screening regimens have also been emerging. This review focuses on three conditions for which consensus screening recommendations have been evolving in response to new evidence: Beckwith-Wiedemann syndrome, Li-Fraumeni syndrome, and constitutional mismatch repair deficiency syndrome. For each condition, recently proposed screening guidelines and relevant evidence are described and potential future directions for improving cancer surveillance practices are anticipated. Also, the implications of several recent studies exploring the psychosocial aspects of screening in these conditions are discussed. SUMMARY: Significant strides have been made in cancer surveillance for children with hereditary cancer predisposition syndromes. A continued emphasis on consensus-driven screening guidelines and collaborative research evaluating the clinical utility of recommended screening methodologies will lead to further improvements in the clinical outcomes of these vulnerable children.


Asunto(s)
Detección Precoz del Cáncer , Síndromes Neoplásicos Hereditarios/diagnóstico , Niño , Humanos
9.
Acta Neuropathol ; 128(4): 583-95, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25022261

RESUMEN

Germ-line RB-1 mutations predispose to pineoblastoma (PinB), but other predisposing genetic factors are not well established. We recently identified a germ-line DICER1 mutation in a child with a PinB. This was accompanied by loss of heterozygosity (LOH) of the wild-type allele within the tumour. We set out to establish the prevalence of DICER1 mutations in an opportunistically ascertained series of PinBs. Twenty-one PinB cases were studied: Eighteen cases had not undergone previous testing for DICER1 mutations; three patients were known carriers of germ-line DICER1 mutations. The eighteen PinBs were sequenced by Sanger and/or Fluidigm-based next-generation sequencing to identify DICER1 mutations in blood gDNA and/or tumour gDNA. Testing for somatic DICER1 mutations was also conducted on one case with a known germ-line DICER1 mutation. From the eighteen PinBs, we identified four deleterious DICER1 mutations, three of which were germ line in origin, and one for which a germ line versus somatic origin could not be determined; in all four, the second allele was also inactivated leading to complete loss of DICER1 protein. No somatic DICER1 RNase IIIb mutations were identified. One PinB arising in a germ-line DICER1 mutation carrier was found to have LOH. This study suggests that germ-line DICER1 mutations make a clinically significant contribution to PinB, establishing DICER1 as an important susceptibility gene for PinB and demonstrates PinB to be a manifestation of a germ-line DICER1 mutation. The means by which the second allele is inactivated may differ from other DICER1-related tumours.


Asunto(s)
Neoplasias Encefálicas/genética , ARN Helicasas DEAD-box/genética , Mutación de Línea Germinal/genética , Glándula Pineal/patología , Pinealoma/genética , Ribonucleasa III/genética , Adolescente , Niño , Preescolar , Análisis Mutacional de ADN , Salud de la Familia , Femenino , Humanos , Lactante , Masculino , Adulto Joven
10.
Clin Cancer Res ; 30(11): 2342-2350, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38573059

RESUMEN

Tumors of the central nervous system (CNS) comprise the second most common group of neoplasms in childhood. The incidence of germline predisposition among children with brain tumors continues to grow as our knowledge on disease etiology increases. Some children with brain tumors may present with nonmalignant phenotypic features of specific syndromes (e.g., nevoid basal cell carcinoma syndrome, neurofibromatosis type 1 and type 2, DICER1 syndrome, and constitutional mismatch-repair deficiency), while others may present with a strong family history of cancer (e.g., Li-Fraumeni syndrome) or with a rare tumor commonly found in the context of germline predisposition (e.g., rhabdoid tumor predisposition syndrome). Approximately 50% of patients with a brain tumor may be the first in a family identified to have a predisposition. The past decade has witnessed a rapid expansion in our molecular understanding of CNS tumors. A significant proportion of CNS tumors are now well characterized and known to harbor specific genetic changes that can be found in the germline. Additional novel predisposition syndromes are also being described. Identification of these germline syndromes in individual patients has not only enabled cascade testing of family members and early tumor surveillance but also increasingly affected cancer management in those patients. Therefore, the AACR Cancer Predisposition Working Group chose to highlight these advances in CNS tumor predisposition and summarize and/or generate surveillance recommendations for established and more recently emerging pediatric brain tumor predisposition syndromes.


Asunto(s)
Neoplasias Encefálicas , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Síndromes Neoplásicos Hereditarios , Humanos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/epidemiología , Neoplasias Encefálicas/diagnóstico , Niño , Síndromes Neoplásicos Hereditarios/genética , Síndromes Neoplásicos Hereditarios/diagnóstico , Síndromes Neoplásicos Hereditarios/epidemiología , Pruebas Genéticas , Guías de Práctica Clínica como Asunto
11.
Pediatr Blood Cancer ; 60(4): 593-8, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23065688

RESUMEN

BACKGROUND: Glutathione S-transferase (GST) enzymes are involved in detoxifying chemotherapy and clearing reactive oxygen species formed by radiation. We explored the relationship between the host GSTP1 105 A > G polymorphism (rs1695), tumor GSTpi protein expression, and clinical outcomes in pediatric medulloblastoma. We hypothesized that the GSTP1 105 G-allele and increased tumor GSTpi expression would be associated with lower progression-free survival and fewer adverse events. PROCEDURE: The study included 106 medulloblastoma/primitive neuroectodermal tumor (PNET) patients seen at Texas Children's Cancer Center. Genotyping was performed using an Illumina HumanOmni1-Quad BeadChip and GSTpi expression was assessed using immunohistochemistry. We used the Kaplan-Meier method for survival analyses and logistic regression for toxicity comparisons. RESULTS: Patients with a GSTP1 105 AG/GG genotype (vs. AA) or who had received high dose craniospinal radiation (≥34 Gy vs. <26 Gy) had a greater risk of requiring hearing aids than their counterparts (OR 4.0, 95% CI 1.2-13.6, and OR 3.1, 95% CI 1.1-8.8, respectively, n = 69). Additionally, there was a statistically significant interaction between these variables. Compared with the lowest risk group (GSTP1 105 AA-low dose radiation), patients with a GSTP1 105 AG/GG genotype who received high dose radiation were 8.4 times more likely to require hearing aids (95% CI 1.4-49.9, p-trend = 0.005, n = 69). When adjusted for age, cumulative cisplatin dose, and amifostine use, the association remained. CONCLUSIONS: The GSTP1 105 G-allele is associated with permanent ototoxicity in pediatric medulloblastoma/PNET and strongly interacts with radiation dose. Patients with this allele should be considered for clinical trials employing radiation dose modifications and cytoprotectant strategies.


Asunto(s)
Neoplasias Cerebelosas/radioterapia , Predisposición Genética a la Enfermedad/genética , Gutatión-S-Transferasa pi/genética , Pérdida Auditiva/etiología , Meduloblastoma/radioterapia , Polimorfismo de Nucleótido Simple , Radioterapia/efectos adversos , Adolescente , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/mortalidad , Niño , Preescolar , Supervivencia sin Enfermedad , Femenino , Genotipo , Humanos , Inmunohistoquímica , Lactante , Estimación de Kaplan-Meier , Masculino , Meduloblastoma/genética , Meduloblastoma/mortalidad , Tumores Neuroectodérmicos Primitivos/genética , Tumores Neuroectodérmicos Primitivos/mortalidad , Tumores Neuroectodérmicos Primitivos/radioterapia
12.
medRxiv ; 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37961416

RESUMEN

Background: Current clinical variant analysis pipelines focus on coding variants and intronic variants within 10-20 bases of an exon-intron boundary that may affect splicing. The impact of newer splicing prediction algorithms combined with in vitro splicing assays on rare variants currently considered Benign/Likely Benign (B/LB) is unknown. Methods: Exome sequencing data from 576 pediatric cancer patients enrolled in the Texas KidsCanSeq study were filtered for intronic or synonymous variants absent from population databases, predicted to alter splicing via SpliceAI (>0.20), and scored as potentially deleterious by CADD (>10.0). Total cellular RNA was extracted from monocytes and RT-PCR products analyzed. Subsequently, rare synonymous or intronic B/LB variants in a subset of genes submitted to ClinVar were similarly evaluated. Variants predicted to lead to a frameshifted splicing product were functionally assessed using an in vitro splicing reporter assay in HEK-293T cells. Results: KidsCanSeq exome data analysis revealed a rare, heterozygous, intronic variant (NM_177438.3(DICER1):c.574-26A>G) predicted by SpliceAI to result in gain of a secondary splice acceptor site. The proband had a personal and family history of pleuropulmonary blastoma consistent with DICER1 syndrome but negative clinical sequencing reports. Proband RNA analysis revealed alternative DICER1 transcripts including the SpliceAI-predicted transcript.Similar bioinformatic analysis of synonymous or intronic B/LB variants (n=31,715) in ClinVar from 61 Mendelian disease genes yielded 18 variants, none of which could be scored by MaxEntScan. Eight of these variants were assessed (DICER1 n=4, CDH1 n=2, PALB2 n=2) using in vitro splice reporter assay and demonstrated abnormal splice products (mean 66%; range 6% to 100%). Available phenotypic information from submitting laboratories demonstrated DICER1 phenotypes in 2 families (1 variant) and breast cancer phenotypes for PALB2 in 3 families (2 variants). Conclusions: Our results demonstrate the power of newer predictive splicing algorithms to highlight rare variants previously considered B/LB in patients with features of hereditary conditions. Incorporation of SpliceAI annotation of existing variant data combined with either direct RNA analysis or in vitro assays has the potential to identify disease-associated variants in patients without a molecular diagnosis.

13.
Cancer Prev Res (Phila) ; 15(10): 645-652, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-36001348

RESUMEN

Germline pathogenic variants in cancer susceptibility genes are identified in up to 18% of all children with cancer. Because pediatric cancer predisposition syndromes (CPS) themselves are rare and underrecognized, there are limited data to guide the diagnosis and management of affected children and at-risk relatives. Furthermore, the care of affected children requires distinct considerations given the early onset of cancers, lifelong risks of additional cancers, and potential late effects of therapy. Herein, we discuss efforts to leverage existing infrastructure, organize experts, and develop a new consortium to optimize care and advance research for children with CPS. A 2016 workshop organized by the American Association for Cancer Research united many experts in childhood cancer predisposition and resulted in publication of multiple consensus guidelines for tumor surveillance. More recently, several of these authors established the Consortium for Childhood Cancer Predisposition (C3P), a multi-institutional collaboration that provides a structure for systematic research in cancer predisposition, screening, and prevention in children. The Consortium intends to work with other cooperative groups to merge longitudinal data from children with CPS throughout the continuum of the cancer risk period, as well as cancer treatment and survivorship care, to optimize overall outcomes.


Asunto(s)
Neoplasias , Niño , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Tamizaje Masivo , Neoplasias/diagnóstico , Neoplasias/genética , Neoplasias/prevención & control
14.
J Pediatr Hematol Oncol ; 33(5): 387-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20829717

RESUMEN

Pediatric oral squamous cell carcinoma is an extremely rare occurence. In our report, we describe a 6-year-old White female with sensorineural hearing loss found to have a Connexin 26 gene mutation who developed a well-differentiated squamous cell carcinoma of the hard palate with metastatic disease to submandibular lymph nodes and the lungs. This association emphasizes the need to consider Connexin 26 gene mutations in children who develop oral squamous cell carcinoma and to monitor for oral squamous cell carcinoma in children with Connexin 26 gene mutations.


Asunto(s)
Carcinoma de Células Escamosas/genética , Conexinas/genética , Neoplasias Orofaríngeas/genética , Neoplasias Palatinas/genética , Carcinoma de Células Escamosas/secundario , Niño , Conexina 26 , Resultado Fatal , Femenino , Pérdida Auditiva Sensorineural/genética , Humanos , Metástasis Linfática , Mutación , Neoplasias Orofaríngeas/secundario , Neoplasias Palatinas/patología
15.
Pediatr Blood Cancer ; 54(7): 1041-4, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20162687

RESUMEN

We describe a male with a large abdominal mass, most likely originating from the liver, with capsule rupture and tumor dissemination into the abdominal cavity. Adherence of the tumor to the diaphragm and lower right colon also were noted. A comprehensive evaluation of the mass revealed no tumor-defining histopathologic, immunocytochemical, ultrastructural, cytogenetic, or translocation features. The malignant tumor was found to have a novel translocation (X;19)(q13;13.3), which has not been reported in small round cell tumors of childhood or adults. The final diagnosis rendered was an undifferentiated small round cell tumor of uncertain cell of origin.


Asunto(s)
Neoplasias Abdominales/genética , Neoplasias Abdominales/patología , Carcinoma de Células Pequeñas/genética , Carcinoma de Células Pequeñas/patología , Cromosomas Humanos Par 19/genética , Cromosomas Humanos X/genética , Neoplasias Abdominales/metabolismo , Carcinoma de Células Pequeñas/metabolismo , Niño , Humanos , Inmunohistoquímica , Masculino , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Translocación Genética
16.
Artículo en Inglés | MEDLINE | ID: mdl-31624068

RESUMEN

Ultra-hypermutation (>100 mutations/Mb) is rare in childhood cancer genomes and has been primarily reported in patients with constitutional mismatch repair deficiency (CMMRD) caused by biallelic germline mismatch repair (MMR) gene mutations. We report a 5-yr-old child with classic clinical features of CMMRD and an ultra-hypermutated medulloblastoma with retained MMR protein expression and absence of germline MMR mutations. Mutational signature analysis of tumor panel sequencing data revealed a canonical DNA polymerase-deficiency-associated signature, prompting further genetic testing that uncovered a germline POLE p.A456P missense variant, which has previously been reported as a recurrent somatic driver mutation in cancers. This represents the earliest known onset of malignancy in a patient with a germline mutation in the POLE proofreading polymerase. The clinical features in this child, virtually indistinguishable from those of CMMRD, suggest that polymerase-proofreading deficiency should be considered in the differential diagnosis of CMMRD patients with retained MMR function.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Colorrectales/genética , ADN Polimerasa II/genética , Meduloblastoma/genética , Síndromes Neoplásicos Hereditarios/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Cerebelosas , Preescolar , Neoplasias Colorrectales/metabolismo , Reparación de la Incompatibilidad de ADN/genética , Análisis Mutacional de ADN , ADN Polimerasa II/metabolismo , Proteínas de Unión al ADN/genética , Femenino , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Células Germinativas/metabolismo , Mutación de Línea Germinal/genética , Humanos , Meduloblastoma/metabolismo , Mutación , Síndromes Neoplásicos Hereditarios/metabolismo , Fenotipo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo
17.
Cancer Epidemiol ; 59: 71-74, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30703618

RESUMEN

INTRODUCTION: Methylation-derived neutrophil-to-lymphocyte ratio (mdNLR) has been identified as a potential prognostic biomarker of outcomes in various cancers. We evaluated the prognostic value of blood-derived mdNLR within a retrospective cohort of pediatric medulloblastoma patients. MATERIALS AND METHODS: DNA methylation was measured in archival peripheral blood samples collected on 56 pediatric medulloblastoma patients. Hazard ratios (HR) and 95% confidence intervals (CI) for the association between mdNLR and survival were evaluated using Cox proportional hazard models. RESULTS: Compared to patients who were alive at last follow-up (n = 43), the mean mdNLR value was slightly higher in deceased patients (n = 13) (12.3 vs. 5.2,P = 0.163). Elevated log-transformed mdNLR was suggestively associated with an increased likelihood of death in unadjusted models (HR=1.43, 95%CI: 0.92-2.22) and significantly associated with mortality in adjusted models (HR=1.61, 95%CI: 1.01-2.58). DISCUSSION: Future work is warranted to investigate the relationship between mdNLR outcomes in specific pediatric medulloblastoma molecular subgroups.


Asunto(s)
Neoplasias Cerebelosas/genética , Metilación de ADN , Meduloblastoma/genética , Adolescente , Neoplasias Cerebelosas/sangre , Neoplasias Cerebelosas/diagnóstico , Neoplasias Cerebelosas/tratamiento farmacológico , Niño , Preescolar , Femenino , Humanos , Recuento de Leucocitos , Linfocitos , Masculino , Meduloblastoma/sangre , Meduloblastoma/diagnóstico , Meduloblastoma/tratamiento farmacológico , Neutrófilos , Proyectos Piloto , Pronóstico , Modelos de Riesgos Proporcionales , Estudios Retrospectivos
18.
Ther Adv Endocrinol Metab ; 9(9): 299-301, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30181857

RESUMEN

Individuals with PTEN Hamartoma Tumor Syndrome (PHTS) are at greatly increased risk for developing well-differentiated thyroid cancer. Specific circumstances in which total thyroidectomies should be considered have not been defined. A 14-year-old macrocephalic female with history of developmental delay and lipoma over her left flank presented with neck swelling and was found have multinodular goiter and auto-immune thyroiditis. Asymptomatic tracheal narrowing was also detected on her initial diagnostic imaging. Later on, she developed positional dyspnea during sleep. Genetic testing revealed a heterozygous pathogenic variant in the PTEN gene (c.463T>A). A total thyroidectomy was performed. In addition to addressing the symptomology in our case, a total thyroidectomy also fortuitously eliminated the thyroid cancer risk. This case spurred us on further to identify specific clinical scenarios where total thyroidectomy may be considered as a true prophylactic measure to manage thyroid cancer risk in PHTS patients.

19.
J Neurosurg Pediatr ; 19(1): 102-107, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27689246

RESUMEN

Neurofibrosarcoma is rare in the pediatric age group. A malignant tumor of the sacrum presents significant challenges, especially if the goals are to resect with wide and clean surgical margins and to achieve acceptable functional outcomes. The authors report a case of this rare tumor affecting the sacrum and sacral nerve roots of a 7-year-old girl and review the role of total hemisacrectomy sparing the contralateral sacral nerve roots and lumbopelvic reconstruction in the treatment of this disease. This patient is, to the best of the authors' knowledge, the youngest to be treated in this manner.


Asunto(s)
Neurofibrosarcoma/cirugía , Huesos Pélvicos/cirugía , Procedimientos de Cirugía Plástica/métodos , Articulación Sacroiliaca/cirugía , Sacro/cirugía , Raíces Nerviosas Espinales/cirugía , Niño , Femenino , Estudios de Seguimiento , Humanos , Neurofibrosarcoma/diagnóstico por imagen , Huesos Pélvicos/diagnóstico por imagen , Articulación Sacroiliaca/diagnóstico por imagen , Sacro/diagnóstico por imagen , Raíces Nerviosas Espinales/diagnóstico por imagen
20.
Neuro Oncol ; 19(10): 1372-1379, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28444219

RESUMEN

BACKGROUND: Ototoxicity is a common adverse side effect of platinum chemotherapy and cranial radiation therapy; however, individual susceptibility is highly variable. Therefore, our objective was to conduct an epigenome-wide association study to identify differentially methylated cytosine-phosphate-guanine (CpG) sites associated with ototoxicity susceptibility among cisplatin-treated pediatric patients with embryonal tumors. METHODS: Samples were collected for a discovery cohort (n = 62) and a replication cohort (n = 18) of medulloblastoma and primitive neuroectodermal tumor patients. Posttreatment audiograms were evaluated using the International Society of Paediatric Oncology (SIOP) Boston Ototoxicity Scale. Genome-wide associations between CpG methylation and ototoxicity were examined using multiple linear regression, controlling for demographic and treatment factors. RESULTS: The mean cumulative dose of cisplatin was 330 mg/m2 and the mean time from end of therapy to the last available audiogram was 6.9 years. In the discovery analysis of 435233 CpG sites, 6 sites were associated with ototoxicity grade (P < 5 × 10-5) after adjusting for confounders. Differential methylation at the top CpG site identified in the discovery cohort (cg14010619, PAK4 gene) was replicated (P = 0.029) and reached genome-wide significance (P = 2.73 × 10-8) in a combined analysis. These findings were robust to a sensitivity analysis evaluating other potential confounders. CONCLUSIONS: We identified and replicated a novel CpG methylation loci (cg14010619) associated with ototoxicity severity. Methylation at cg14010619 may modify PAK4 activity, which has been implicated in cisplatin resistance in malignant cell lines.


Asunto(s)
Neoplasias Cerebelosas/terapia , Cisplatino/uso terapéutico , Metilación de ADN/fisiología , Meduloblastoma/terapia , Quinasas p21 Activadas/metabolismo , Adolescente , Antineoplásicos/uso terapéutico , Niño , Preescolar , Estudios de Cohortes , Femenino , Pérdida Auditiva/inducido químicamente , Humanos , Masculino , Neoplasias de Células Germinales y Embrionarias/tratamiento farmacológico , Adulto Joven , Quinasas p21 Activadas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA