Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Clin Cancer Res ; 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38573059

RESUMO

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 aetiology increases. Some children with brain tumors may present with non-malignant 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 increasingly has also impacted 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.

2.
Cancer ; 130(3): 467-475, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-37788149

RESUMO

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.


Assuntos
Síndrome de Beckwith-Wiedemann , Neoplasias Renais , Neoplasias Hepáticas , Tumor de Wilms , Lactente , Criança , Humanos , Incidência , Síndrome de Beckwith-Wiedemann/complicações , Síndrome de Beckwith-Wiedemann/epidemiologia , Síndrome de Beckwith-Wiedemann/genética , Tumor de Wilms/epidemiologia , Neoplasias Renais/complicações , Neoplasias Hepáticas/complicações
3.
medRxiv ; 2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37961416

RESUMO

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.

4.
Cancer Prev Res (Phila) ; 15(10): 645-652, 2022 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-36001348

RESUMO

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.


Assuntos
Neoplasias , Criança , Predisposição Genética para Doença , Genótipo , Humanos , Programas de Rastreamento , Neoplasias/diagnóstico , Neoplasias/genética , Neoplasias/prevenção & controle
5.
Pediatr Blood Cancer ; 68(10): e29178, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34133064

RESUMO

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.


Assuntos
Gadolínio , Glioma , Encéfalo/diagnóstico por imagem , Criança , Meios de Contraste , Glioma/diagnóstico por imagem , Humanos , Imageamento por Ressonância Magnética , Estudos Retrospectivos
6.
Pediatr Blood Cancer ; 67(6): e28258, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32196895

RESUMO

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.


Assuntos
Neoplasias Gastrointestinais/patologia , Mutação , Neovascularização Patológica/complicações , PTEN Fosfo-Hidrolase/genética , Neoplasias da Glândula Tireoide/patologia , Malformações Vasculares/complicações , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Seguimentos , Neoplasias Gastrointestinais/etiologia , Neoplasias Gastrointestinais/genética , Predisposição Genética para Doença , Genótipo , Humanos , Lactente , Recém-Nascido , Masculino , Fenótipo , Prognóstico , Estudos Retrospectivos , Neoplasias da Glândula Tireoide/etiologia , Neoplasias da Glândula Tireoide/genética , Adulto Jovem
7.
Artigo em Inglês | MEDLINE | ID: mdl-31624068

RESUMO

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.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Colorretais/genética , DNA Polimerase II/genética , Meduloblastoma/genética , Síndromes Neoplásicas Hereditárias/genética , Proteínas de Ligação a Poli-ADP-Ribose/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Cerebelares , Pré-Escolar , Neoplasias Colorretais/metabolismo , Reparo de Erro de Pareamento de DNA/genética , Análise Mutacional de DNA , DNA Polimerase II/metabolismo , Proteínas de Ligação a DNA/genética , Feminino , Predisposição Genética para Doença , Testes Genéticos , Células Germinativas/metabolismo , Mutação em Linhagem Germinativa/genética , Humanos , Meduloblastoma/metabolismo , Mutação , Síndromes Neoplásicas Hereditárias/metabolismo , Fenótipo , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo
8.
Cancer Epidemiol ; 59: 71-74, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30703618

RESUMO

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.


Assuntos
Neoplasias Cerebelares/genética , Metilação de DNA , Meduloblastoma/genética , Adolescente , Neoplasias Cerebelares/sangue , Neoplasias Cerebelares/diagnóstico , Neoplasias Cerebelares/tratamento farmacológico , Criança , Pré-Escolar , Feminino , Humanos , Contagem de Leucócitos , Linfócitos , Masculino , Meduloblastoma/sangue , Meduloblastoma/diagnóstico , Meduloblastoma/tratamento farmacológico , Neutrófilos , Projetos Piloto , Prognóstico , Modelos de Riscos Proporcionais , Estudos Retrospectivos
9.
Curr Opin Pediatr ; 31(1): 41-47, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30531401

RESUMO

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.


Assuntos
Detecção Precoce de Câncer , Síndromes Neoplásicas Hereditárias/diagnóstico , Criança , Humanos
10.
Ther Adv Endocrinol Metab ; 9(9): 299-301, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30181857

RESUMO

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.

11.
JAMA Oncol ; 3(12): 1634-1639, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28772291

RESUMO

Importance: Guidelines for clinical management in Li-Fraumeni syndrome, a multiple-organ cancer predisposition condition, are limited. Whole-body magnetic resonance imaging (WBMRI) may play a role in surveillance of this high-risk population. Objective: To assess the clinical utility of WBMRI in germline TP53 mutation carriers at baseline. Data Sources: Clinical and research surveillance cohorts were identified through the Li-Fraumeni Exploration Research Consortium. Study Selection: Cohorts that incorporated WBMRI for individuals with germline TP53 mutations from January 1, 2004, through October 1, 2016, were included. Data Extraction and Synthesis: Data were extracted by investigators from each cohort independently and synthesized by 2 investigators. Random-effects meta-analysis methods were used to estimate proportions. Main Outcomes and Measures: The proportions of participants at baseline in whom a lesion was detected that required follow-up and in whom a new primary malignant neoplasm was detected. Results: A total of 578 participants (376 female [65.1%] and 202 male [34.9%]; mean [SD] age, 33.2 [17.1] years) from 13 cohorts in 6 countries were included in the analysis. Two hundred twenty-five lesions requiring clinical follow-up were detected by WBMRI in 173 participants. Sixty-one lesions were diagnosed in 54 individuals as benign or malignant neoplasms. Overall, 42 cancers were identified in 39 individuals, with 35 new localized cancers treated with curative intent. The overall estimated detection rate for new, localized primary cancers was 7% (95% CI, 5%-9%). Conclusions and Relevance: These data suggest clinical utility of baseline WBMRI in TP53 germline mutation carriers and may form an integral part of baseline clinical risk management in this high-risk population.


Assuntos
Síndrome de Li-Fraumeni/diagnóstico por imagem , Síndrome de Li-Fraumeni/epidemiologia , Proteína Supressora de Tumor p53/genética , Imagem Corporal Total/métodos , Adolescente , Adulto , Diagnóstico Diferencial , Detecção Precoce de Câncer , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Mutação , Vigilância da População , Guias de Prática Clínica como Assunto , Adulto Jovem
12.
Clin Cancer Res ; 23(13): e123-e132, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28674121

RESUMO

Children and adolescents who present with neuroendocrine tumors are at extremely high likelihood of having an underlying germline predisposition for the multiple endocrine neoplasia (MEN) syndromes, including MEN1, MEN2A and MEN2B, MEN4, and hyperparathyroid-jaw tumor (HPT-JT) syndromes. Each of these autosomal dominant syndromes results from a specific germline mutation in unique genes: MEN1 is due to pathogenic MEN1 variants (11q13), MEN2A and MEN2B are due to pathogenic RET variants (10q11.21), MEN4 is due to pathogenic CDKN1B variants (12p13.1), and the HPT-JT syndrome is due to pathogenic CDC73 variants (1q25). Although each of these genetic syndromes share the presence of neuroendocrine tumors, each syndrome has a slightly different tumor spectrum with specific surveillance recommendations based upon tumor penetrance, including the age and location for which specific tumor types most commonly present. Although the recommended surveillance strategies for each syndrome contain similar approaches, important differences do exist among them. Therefore, it is important for caregivers of children and adolescents with these syndromes to become familiar with the unique diagnostic criteria for each syndrome, and also to be aware of the specific tumor screening and prophylactic surgery recommendations for each syndrome. Clin Cancer Res; 23(13); e123-e32. ©2017 AACRSee all articles in the online-only CCR Pediatric Oncology Series.


Assuntos
Adenoma/genética , Fibroma/genética , Hiperparatireoidismo/genética , Neoplasias Maxilomandibulares/genética , Neoplasia Endócrina Múltipla/genética , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Supressoras de Tumor/genética , Adenoma/diagnóstico , Adenoma/epidemiologia , Adolescente , Criança , Fibroma/diagnóstico , Fibroma/epidemiologia , Predisposição Genética para Doença , Mutação em Linhagem Germinativa/genética , Humanos , Hiperparatireoidismo/diagnóstico , Hiperparatireoidismo/epidemiologia , Neoplasias Maxilomandibulares/diagnóstico , Neoplasias Maxilomandibulares/epidemiologia , Neoplasia Endócrina Múltipla/diagnóstico , Neoplasia Endócrina Múltipla/epidemiologia , Neoplasia Endócrina Múltipla Tipo 2b/diagnóstico , Neoplasia Endócrina Múltipla Tipo 2b/epidemiologia , Neoplasia Endócrina Múltipla Tipo 2b/genética , Fatores de Risco
13.
Clin Cancer Res ; 23(12): e76-e82, 2017 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-28620008

RESUMO

PTEN hamartoma tumor syndrome (PHTS), DICER1 syndrome, and hereditary leiomyomatosis and renal cell cancer (HLRCC) syndrome are pleiotropic tumor predisposition syndromes that include benign and malignant neoplasms affecting adults and children. PHTS includes several disorders with shared and distinct clinical features. These are associated with elevated lifetime risk of breast, thyroid, endometrial, colorectal, and renal cancers as well as melanoma. Thyroid cancer represents the predominant cancer risk under age 20 years. DICER1 syndrome includes risk for pleuropulmonary blastoma, cystic nephroma, ovarian sex cord-stromal tumors, and multinodular goiter and thyroid carcinoma as well as brain tumors including pineoblastoma and pituitary blastoma. Individuals with HLRCC may develop multiple cutaneous and uterine leiomyomas, and they have an elevated risk of renal cell carcinoma. For each of these syndromes, a summary of the key syndromic features is provided, the underlying genetic events are discussed, and specific screening is recommended. Clin Cancer Res; 23(12); e76-e82. ©2017 AACRSee all articles in the online-only CCR Pediatric Oncology Series.


Assuntos
RNA Helicases DEAD-box/genética , Fumarato Hidratase/genética , Síndrome do Hamartoma Múltiplo/genética , Leiomiomatose/genética , Síndromes Neoplásicas Hereditárias/genética , PTEN Fosfo-Hidrolase/genética , Ribonuclease III/genética , Neoplasias Cutâneas/genética , Neoplasias Uterinas/genética , Criança , Detecção Precoce de Câncer , Síndrome do Hamartoma Múltiplo/epidemiologia , Síndrome do Hamartoma Múltiplo/patologia , Humanos , Leiomiomatose/epidemiologia , Leiomiomatose/patologia , Síndromes Neoplásicas Hereditárias/epidemiologia , Síndromes Neoplásicas Hereditárias/patologia , Fatores de Risco , Neoplasias Cutâneas/epidemiologia , Neoplasias Cutâneas/patologia , Neoplasias Uterinas/epidemiologia , Neoplasias Uterinas/patologia
14.
Clin Cancer Res ; 23(12): e68-e75, 2017 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-28620007

RESUMO

Von Hippel-Lindau disease (vHL) is a hereditary tumor predisposition syndrome that places affected individuals at risk for multiple tumors, which are predominantly benign and generally occur in the central nervous system or abdomen. Although the majority of tumors occur in adults, children and adolescents with the condition develop a significant proportion of vHL manifestations and are vulnerable to delayed tumor detection and their sequelae. Although multiple tumor screening paradigms are currently being utilized for patients with vHL, surveillance should be reassessed as the available relevant clinical information continues to expand. We propose a new vHL screening paradigm similar to existing approaches, with important modifications for some tumor types, placing an emphasis on risks in childhood. This includes advancement in the timing of surveillance initiation and increased frequency of screening evaluations. Another neuroendocrine-related familial condition is the rapidly expanding hereditary paraganglioma and pheochromocytoma syndrome (HPP). The tumor spectrum for patients with HPP syndrome includes paragangliomas, pheochromocytomas, renal cancer, and gastrointestinal stromal tumors. The majority of patients with HPP syndrome harbor an underlying variant in one of the SHDx genes (SDHA, SDHB, SDHC, SDHD, SDHA, and SDHAF2), although other genes also have been described (MAX and TMEM127). Annual screening for elevated plasma or urine markers along with complete blood count and biennial whole-body MRI accompanied by focal neck MRI is recommended for older children and adults with HPP syndrome to detect tumors early and to decrease morbidity and mortality from HPP-related tumors. Clin Cancer Res; 23(12); e68-e75. ©2017 AACRSee all articles in the online-only CCR Pediatric Oncology Series.


Assuntos
Neoplasias das Glândulas Suprarrenais/diagnóstico , Paraganglioma/diagnóstico , Feocromocitoma/diagnóstico , Doença de von Hippel-Lindau/diagnóstico , Neoplasias das Glândulas Suprarrenais/epidemiologia , Neoplasias das Glândulas Suprarrenais/patologia , Detecção Precoce de Câncer , Humanos , Mutação , Proteínas de Neoplasias/genética , Paraganglioma/epidemiologia , Paraganglioma/genética , Paraganglioma/patologia , Feocromocitoma/epidemiologia , Feocromocitoma/genética , Feocromocitoma/patologia , Fatores de Risco , Doença de von Hippel-Lindau/epidemiologia , Doença de von Hippel-Lindau/genética , Doença de von Hippel-Lindau/patologia
15.
Neuro Oncol ; 19(10): 1372-1379, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-28444219

RESUMO

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.


Assuntos
Neoplasias Cerebelares/terapia , Cisplatino/uso terapêutico , Metilação de DNA/fisiologia , Meduloblastoma/terapia , Quinases Ativadas por p21/metabolismo , Adolescente , Antineoplásicos/uso terapêutico , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Perda Auditiva/induzido quimicamente , Humanos , Masculino , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Adulto Jovem , Quinases Ativadas por p21/genética
16.
Cancer Epidemiol ; 44: 161-166, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27607585

RESUMO

AIM: Medulloblastoma is the most frequent malignant pediatric brain tumor. While survival rates have improved due to multimodal treatment including cisplatin-based chemotherapy, there are few prognostic factors for adverse treatment outcomes. Notably, genes involved in the nucleotide excision repair pathway, including ERCC2, have been implicated in cisplatin sensitivity in other cancers. Therefore, this study evaluated the role of ERCC2 DNA methylation profiles on pediatric medulloblastoma survival. METHODS: The study population included 71 medulloblastoma patients (age <18years at diagnosis) and recruited from Texas Children's Cancer Center between 2004 and 2009. DNA methylation profiles were generated from peripheral blood samples using the Illumina Infinium Human Methylation 450 Beadchip. Sixteen ERCC2-associated CpG sites were evaluated in this analysis. Multivariable regression models were used to determine the adjusted association between DNA methylation and survival. Cox regression and Kaplan-Meier curves were used to compare 5-year overall survival between hyper- and hypo-methylation at each CpG site. RESULTS: In total, 12.7% (n=9) of the patient population died within five years of diagnosis. In our population, methylation of the cg02257300 probe (Hazard Ratio=9.33; 95% Confidence Interval: 1.17-74.64) was associated with death (log-rank p=0.01). This association remained suggestive after correcting for multiple comparisons (FDR p<0.2). No other ERCC2-associated CpG site was associated with survival in this population of pediatric medulloblastoma patients. CONCLUSION: These findings provide the first evidence that DNA methylation within the promoter region of the ERCC2 gene may be associated with survival in pediatric medulloblastoma. If confirmed in future studies, this information may lead to improved risk stratification or promote the development of novel, targeted therapeutics.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Cerebelares/mortalidade , Metilação de DNA , DNA de Neoplasias/genética , Meduloblastoma/mortalidade , Proteína Grupo D do Xeroderma Pigmentoso/genética , Adolescente , Neoplasias Cerebelares/genética , Neoplasias Cerebelares/patologia , Criança , Pré-Escolar , Ilhas de CpG/genética , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Meduloblastoma/genética , Meduloblastoma/patologia , Regiões Promotoras Genéticas/genética , Texas
17.
Acta Neuropathol ; 128(4): 583-95, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25022261

RESUMO

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.


Assuntos
Neoplasias Encefálicas/genética , RNA Helicases DEAD-box/genética , Mutação em Linhagem Germinativa/genética , Glândula Pineal/patologia , Pinealoma/genética , Ribonuclease III/genética , Adolescente , Criança , Pré-Escolar , Análise Mutacional de DNA , Saúde da Família , Feminino , Humanos , Lactente , Masculino , Adulto Jovem
18.
Cell Signal ; 22(3): 386-94, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19874889

RESUMO

IkappaBalpha serves as a central anchoring molecule in the sequestration of NF-kappaB transcription factor in the cytoplasm. Ubiquitination-mediated IkappaBalpha degradation immediately precedes and is required for NF-kappaB nuclear translocation and activation. However, the precise mechanism for the deubiquitination of IkappaBalpha is still not fully understood. Using a proteomic approach, we have identified Ubiquitin Specific Peptidase 11 (USP11) as an IkappaBalpha associated deubiquitinase. Overexpression of USP11 inhibits IkappaBalpha ubiquitination. Recombinant USP11 catalyzes deubiquitination of IkappaBalpha in vitro. Moreover, knockdown of USP11 expression enhances TNFalpha-induced IkappaBalpha ubiquitination and NF-kappaB activation. These data demonstrate that USP11 plays an important role in the downregulation of TNFalpha-mediated NF-kappaB activation through modulating IkappaBalpha stability. In addition, overexpression of a catalytically inactive USP11 mutant partially inhibits TNFalpha- and IKKbeta-induced NF-kappaB activation, suggesting that USP11 also exerts a non-catalytic function in its negative regulation of TNFalpha-mediated NF-kappaB activation. Thus, IkappaBalpha ubiquitination and deubiquitination processes function as a Yin-Yang regulatory mechanism on TNFalpha-induced NF-kappaB activation.


Assuntos
Quinase I-kappa B/metabolismo , NF-kappa B/metabolismo , Tioléster Hidrolases/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Linhagem Celular , Regulação para Baixo , Células HeLa , Humanos , Interleucina-6/metabolismo , Tioléster Hidrolases/deficiência , Tioléster Hidrolases/genética , Ubiquitinação , Yin-Yang
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...