Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 259
Filter
Add more filters

Publication year range
1.
Cancer Causes Control ; 34(11): 1005-1015, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37421504

ABSTRACT

PURPOSE: Studies report mixed findings regarding the association of breastfeeding with childhood brain tumors (CBT), the leading causes of cancer-related mortality in young people. Our objective was to determine whether breastfeeding is associated with CBT incidence. METHODS: We pooled data on N = 2610 cases with CBT (including 697 cases with astrocytoma, 447 cases with medulloblastoma/primitive neuroectodermal tumor [PNET], 167 cases with ependymoma) and N = 8128 age- and sex-matched controls in the Childhood Cancer and Leukemia International Consortium. We computed unconditional logistic regression models to estimate the odds ratio (OR) and 95% confidence interval (CI) of CBT, astrocytoma, medulloblastoma/PNET, and ependymoma according to breastfeeding status, adjusting for study, sex, mode of delivery, birthweight, age at diagnosis/interview, maternal age at delivery, maternal educational attainment, and maternal race/ethnicity. We evaluated any breastfeeding versus none and breastfeeding ≥ 6 months versus none. We subsequently performed random effects meta-analysis to confirm our findings, identify potential sources of heterogeneity, and evaluate for outliers or influential studies. RESULTS: Breastfeeding was reported by 64.8% of control mothers and 64.5% of case mothers and was not associated with CBT (OR 1.04, 95% CI 0.94-1.15), astrocytoma (OR 1.01, 95% CI 0.87-1.17), medulloblastoma/PNET (OR 1.11, 95% CI 0.93-1.32), or ependymoma (OR 1.06, 95% CI 0.81-1.40). Results were similar when we restricted to breastfeeding ≥ 6 months and in meta-analyses. CONCLUSION: Our data suggest that breastfeeding does not protect against CBT.


Subject(s)
Astrocytoma , Brain Neoplasms , Cerebellar Neoplasms , Ependymoma , Leukemia , Medulloblastoma , Neuroectodermal Tumors, Primitive , Child , Female , Humans , Infant , Astrocytoma/epidemiology , Astrocytoma/etiology , Brain Neoplasms/epidemiology , Brain Neoplasms/etiology , Breast Feeding , Case-Control Studies , Ependymoma/epidemiology , Leukemia/epidemiology , Medulloblastoma/epidemiology , Neuroectodermal Tumors, Primitive/epidemiology , Risk Factors , Male
2.
Brain ; 143(1): 131-149, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31834371

ABSTRACT

Tuberous sclerosis complex (TSC) is an autosomal dominantly inherited neurocutaneous disorder caused by inactivating mutations in TSC1 or TSC2, key regulators of the mechanistic target of rapamycin complex 1 (mTORC1) pathway. In the CNS, TSC is characterized by cortical tubers, subependymal nodules and subependymal giant cell astrocytomas (SEGAs). SEGAs may lead to impaired circulation of CSF resulting in hydrocephalus and raised intracranial pressure in patients with TSC. Currently, surgical resection and mTORC1 inhibitors are the recommended treatment options for patients with SEGA. In the present study, high-throughput RNA-sequencing (SEGAs n = 19, periventricular control n = 8) was used in combination with computational approaches to unravel the complexity of SEGA development. We identified 9400 mRNAs and 94 microRNAs differentially expressed in SEGAs compared to control tissue. The SEGA transcriptome profile was enriched for the mitogen-activated protein kinase (MAPK) pathway, a major regulator of cell proliferation and survival. Analysis at the protein level confirmed that extracellular signal-regulated kinase (ERK) is activated in SEGAs. Subsequently, the inhibition of ERK independently of mTORC1 blockade decreased efficiently the proliferation of primary patient-derived SEGA cultures. Furthermore, we found that LAMTOR1, LAMTOR2, LAMTOR3, LAMTOR4 and LAMTOR5 were overexpressed at both gene and protein levels in SEGA compared to control tissue. Taken together LAMTOR1-5 can form a complex, known as the 'Ragulator' complex, which is known to activate both mTORC1 and MAPK/ERK pathways. Overall, this study shows that the MAPK/ERK pathway could be used as a target for treatment independent of, or in combination with mTORC1 inhibitors for TSC patients. Moreover, our study provides initial evidence of a possible link between the constitutive activated mTORC1 pathway and a secondary driver pathway of tumour growth.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , MAP Kinase Signaling System/genetics , MicroRNAs/metabolism , RNA, Messenger/metabolism , Tuberous Sclerosis/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adolescent , Adult , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytoma/etiology , Astrocytoma/metabolism , Brain Neoplasms/complications , Brain Neoplasms/metabolism , Butadienes/pharmacology , Child , Child, Preschool , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Profiling , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , High-Throughput Nucleotide Sequencing , Humans , Infant , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Nitriles/pharmacology , RNA-Seq , Sequence Analysis, RNA , Tuberous Sclerosis/complications , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/genetics , Tumor Cells, Cultured , Young Adult
3.
Pediatr Radiol ; 50(10): 1397-1408, 2020 09.
Article in English | MEDLINE | ID: mdl-32671416

ABSTRACT

BACKGROUND: Subependymal giant cell astrocytomas (SEGAs) arise in 10-26% of tuberous sclerosis complex (TSC) patients. SEGAs cause obstructive hydrocephalus and increase morbi-mortality. It is recommended that TSC patients be followed with contrast enhanced magnetic resonance imaging (CE-MRI), but repetitive use of gadolinium-based contrast-agents (GBCAs) may cause organ deposits. OBJECTIVE: To compare the diagnostic performances of non-CE- and CE-MRI to differentiate SEGAs from subependymal nodules in TSC patients during follow-up. MATERIALS AND METHODS: Thirty-five TSC patients (median age: 2.4 years) were enrolled in this retrospective single-center study from September 2007 to January 2019. Inclusion criteria were a certain diagnosis of TSC and at least three follow-up brain MRIs with GBCA injection. Two consecutive MRI scans per patient were selected and anonymized. Three radiologists performed a blinded review of non-enhanced and enhanced MRI sequences during different sessions. The diagnostic performances were compared (sensitivity, specificity, positive/negative predictive values, accuracy, inter/intra-observer agreements). RESULTS: The accuracies for detecting SEGAs were good and similar between the non-enhanced and enhanced MRI sequences. The sensitivity and specificity of non-CE-MRI to diagnose SEGA ranged from 75% to 100% and from 94% to 100%, respectively. The differences in numbers of false-positive and false-negative patients between non-CE- and CE-MRI never exceeded one case. Nodules size >10 mm, location near the Monro foramen, hydrocephalus and modifications between two consecutive MRI scans were significantly associated with the diagnosis of SEGA for the three readers (all P-values <0.05). Inter- and intra-observer agreements were also excellent for non-enhanced and enhanced MRI sequences (kappa=0.85-1 and 0.81-0.93, respectively). CONCLUSION: The performances of non-enhanced and enhanced MRI sequences are comparable for detecting SEGAs, questioning the need for systematic GBCA injections for TSC patients.


Subject(s)
Astrocytoma/diagnostic imaging , Brain Neoplasms/diagnostic imaging , Contrast Media/administration & dosage , Magnetic Resonance Imaging/methods , Meglumine/administration & dosage , Organometallic Compounds/administration & dosage , Tuberous Sclerosis/complications , Astrocytoma/etiology , Brain Neoplasms/etiology , Child , Child, Preschool , Female , Humans , Male , Predictive Value of Tests , Retrospective Studies , Sensitivity and Specificity
4.
Int J Mol Sci ; 21(23)2020 Nov 30.
Article in English | MEDLINE | ID: mdl-33266110

ABSTRACT

Astrocytomas and, in particular, their most severe form, glioblastoma, are the most aggressive primary brain tumors and those with the poorest vital prognosis. Standard treatment only slightly improves patient survival. Therefore, new therapies are needed. Very few risk factors have been clearly identified but many epidemiological studies have reported a higher incidence in men than women with a sex ratio of 1:4. Based on these observations, it has been proposed that the neurosteroids and especially the estrogens found in higher concentrations in women's brains could, in part, explain this difference. Estrogens can bind to nuclear or membrane receptors and potentially stimulate many different interconnected signaling pathways. The study of these receptors is even more complex since many isoforms are produced from each estrogen receptor encoding gene through alternative promoter usage or splicing, with each of them potentially having a specific role in the cell. The purpose of this review is to discuss recent data supporting the involvement of steroids during gliomagenesis and to focus on the potential neuroprotective role as well as the mechanisms of action of estrogens in gliomas.


Subject(s)
Astrocytoma/pathology , Hormones/metabolism , Animals , Astrocytoma/classification , Astrocytoma/etiology , Astrocytoma/therapy , Disease Progression , Female , Humans , Male , Models, Biological , Receptors, Steroid/metabolism , Sex Characteristics
5.
Int J Mol Sci ; 21(19)2020 Oct 07.
Article in English | MEDLINE | ID: mdl-33036421

ABSTRACT

Glioma is a lethal central nervous system tumor with poor patient survival prognosis. Because of the molecular heterogeneity, it is a challenge to precisely determine the type of the tumor and to choose the most effective treatment. Therefore, novel biomarkers are essential to improve the diagnosis and prognosis of glioma tumors. Class 3 semaphorin proteins (SEMA3) play an important role in tumor biology. SEMA3 transduce their signals by using neuropilin and plexin receptors, which functionally interact with the vascular endothelial growth factor-mediated signaling pathways. Therefore, the aim of this study was to explore the potential of SEMA3 signaling molecules for prognosis of glioma patient survival. The quantitative real-time PCR method was used to evaluate mRNA expression of SEMA3(A-G), neuropilins (NRP1 and NRP2), plexins (PLXNA2 and PLXND1), cadherins (CDH1 and CDH2), integrins (ITGB1, ITGB3, ITGA5, and ITGAV), VEGFA and KDR genes in 59 II-IV grade glioma tissues. Seven genes significantly associated with patient overall survival were used for multi-biomarker construction, which showed 64%, 75%, and 68% of accuracy of predicting the survival of 1-, 2-, and 3-year glioma patients, respectively. The results suggest that the seven-gene signature could serve as a novel multi-biomarker for more accurate prognosis of a glioma patient's outcome.


Subject(s)
Biomarkers , Glioma/metabolism , Glioma/mortality , Semaphorin-3A/metabolism , Signal Transduction , Aged , Aged, 80 and over , Alleles , Astrocytoma/etiology , Astrocytoma/metabolism , Astrocytoma/mortality , Astrocytoma/pathology , Female , Gene Expression Regulation, Neoplastic , Glioma/etiology , Glioma/pathology , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Mutation , Neoplasm Grading , Prognosis , ROC Curve
6.
J Eur Acad Dermatol Venereol ; 32(10): 1796-1803, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29569806

ABSTRACT

BACKGROUND: Tuberous sclerosis complex (TSC) is a genetic disorder associated with tumour growth in various organs, including the brain, kidneys, heart and skin. Cutaneous lesions are prevalent manifestations of TSC, occurring in up to 90% of patients. Oral mammalian target of rapamycin inhibitors, such as everolimus, is believed to be effective for treatment of TSC-associated lesions because they act on the underlying disease pathophysiology. OBJECTIVE: We evaluated the long-term effect of oral everolimus on TSC-associated skin lesions as a secondary objective in the phase III studies EXIST-1 (NCT00789828) and EXIST-2 (NCT00790400) after approximately 4 years of treatment. MATERIALS AND METHODS: Everolimus was dosed 4.5 mg/m2 /day (titrated to trough 5-15 ng/mL) in patients with TSC-associated subependymal giant cell astrocytoma in EXIST-1, and 10 mg/day initially in adult patients with TSC- or sporadic lymphangioleiomyomatosis-associated renal angiomyolipoma in EXIST-2. Following positive results from the core phase, remaining patients were offered open-label everolimus in an extension. Skin lesion response rate was the proportion of patients achieving complete or partial clinical response. RESULTS: A total of 105 patients in EXIST-1 and 107 in EXIST-2 received everolimus and had ≥1 skin lesion at baseline. Skin lesion response rate (95% confidence interval) was 58.1% (48.1-67.7%) in EXIST-1 and 68.2% (58.5-76.9%) in EXIST-2; most were partial responses. At week 192 (EXIST-1: n = 55; EXIST-2: n = 56), 69% and 66% had a response. Most common drug-related adverse event was stomatitis (41-45%). CONCLUSION: Oral everolimus improved TSC-related skin lesions, with responses sustained over 4 years of treatment in EXIST-1 and EXIST-2.


Subject(s)
Angiomyolipoma/drug therapy , Antineoplastic Agents/therapeutic use , Astrocytoma/drug therapy , Central Nervous System Neoplasms/drug therapy , Everolimus/therapeutic use , Kidney Neoplasms/drug therapy , Skin Neoplasms/drug therapy , Tuberous Sclerosis/drug therapy , Adolescent , Adult , Angiomyolipoma/etiology , Antineoplastic Agents/adverse effects , Astrocytoma/etiology , Central Nervous System Neoplasms/etiology , Child , Child, Preschool , Everolimus/adverse effects , Female , Humans , Infant , Kidney Neoplasms/etiology , Male , Middle Aged , Skin Neoplasms/etiology , Tuberous Sclerosis/complications , Young Adult
7.
Childs Nerv Syst ; 33(4): 601-607, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28074282

ABSTRACT

INTRODUCTION: Tuberous sclerosis is associated with three central nervous system pathologies: cortical/subcortical tubers, subependymal nodules (SENs), and subependymal giant cell astrocytomas (SEGAs). Tubers are associated with epilepsy, which is often medication-resistant and often leads to resective surgery. Recently, mammalian target of rapamycin inhibitors (mTORi) have been shown to be effective reducing seizure burden in some patients with tuberous sclerosis complex (TSC)-related refractory epilepsy. mTORi have also been shown to be an alternative for surgery treating SEGAs. We describe several cases of resected tubers that contained SEGA tissue without an intraventricular SEGA. METHODS: After institutional review board (IRB) protocol approval, we retrospectively reviewed the surgical-pathological data for all TSC patients who underwent cortical resections for treatment of refractory epilepsy at NYU Langone Medical Center and Tel Aviv Medical Center between 2003 and 2013. Data included demographics, epilepsy type, MRI characteristics, epilepsy outcome, and histopathological staining. RESULTS: We reviewed cortical resections from 75 patients with complete pathological studies. In three patients, cortical lesions demonstrated histopathological findings consistent with a SEGA within the resected tuber tissue, with no intraventricular SEGA. All lesions were cortically based and none had any intraventricular extension. No patient had been treated before surgery with an mTORi. Two of the three patients remain Engel grade I-II. All lesions stained positive for glial fibrillary acidic protein (GFAP), synaptophysin, and neuronal nuclear antigen (NeuN). CONCLUSION: This is the first description of cortical tubers harboring SEGA tissue. This observation though preliminary may suggest a subgroup of patients with intractable epilepsy in whom mTORi may be considered before surgical intervention.


Subject(s)
Astrocytoma/etiology , Brain Neoplasms/etiology , Cerebral Cortex/diagnostic imaging , Tuberous Sclerosis/complications , Astrocytoma/diagnostic imaging , Astrocytoma/therapy , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/therapy , Cerebral Cortex/metabolism , Child, Preschool , Cytokines/metabolism , Epilepsy/diagnostic imaging , Epilepsy/etiology , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Infant , Ki-67 Antigen/metabolism , Magnetic Resonance Imaging , Male , Neurosurgical Procedures , Phosphopyruvate Hydratase/metabolism , Retrospective Studies , Tomography Scanners, X-Ray Computed , Tuberous Sclerosis/diagnostic imaging , Tuberous Sclerosis/therapy
8.
Ann Neurol ; 78(6): 929-38, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26381530

ABSTRACT

OBJECTIVE: To analyze the cumulative efficacy and safety of everolimus in treating subependymal giant cell astrocytomas (SEGA) associated with tuberous sclerosis complex (TSC) from an open-label phase II study (NCT00411619). Updated data became available from the conclusion of the extension phase and are presented in this ≥5-year analysis. METHODS: Patients aged ≥ 3 years with a definite diagnosis of TSC and increasing SEGA lesion size (≥2 magnetic resonance imaging scans) received everolimus starting at 3mg/m(2) /day (titrated to target blood trough levels of 5-15ng/ml). The primary efficacy endpoint was reduction from baseline in primary SEGA volume. RESULTS: As of the study completion date (January 28, 2014), 22 of 28 (78.6%) initially enrolled patients finished the study per protocol. Median (range) duration of exposure to everolimus was 67.8 (4.7-83.2) months; 12 (52.2%) and 14 (60.9%) of 23 patients experienced SEGA volume reductions of ≥50% and ≥30% relative to baseline, respectively, after 60 months of treatment. The proportion of patients experiencing daily seizures was reduced from 7 of 26 (26.9%) patients at baseline to 2 of 18 (11.1%) patients at month 60. Most commonly reported adverse events (AEs) were upper respiratory tract infection and stomatitis of mostly grade 1 or 2 severity. No patient discontinued treatment due to AEs. The frequency of emergence of most AEs decreased over the course of the study. INTERPRETATION: Everolimus continues to demonstrate a sustained effect on SEGA tumor reduction over ≥5 years of treatment. Everolimus remained well-tolerated, and no new safety concerns were noted.


Subject(s)
Antineoplastic Agents/pharmacology , Astrocytoma/drug therapy , Brain Neoplasms/drug therapy , Everolimus/pharmacology , Outcome Assessment, Health Care , Tuberous Sclerosis/complications , Adolescent , Adult , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Astrocytoma/etiology , Brain Neoplasms/etiology , Child , Child, Preschool , Everolimus/administration & dosage , Everolimus/adverse effects , Female , Follow-Up Studies , Humans , Male , Young Adult
9.
Am J Med Genet A ; 170(6): 1455-61, 2016 06.
Article in English | MEDLINE | ID: mdl-26992069

ABSTRACT

Although optic pathway gliomas are the most common brain tumors associated with neurofibromatosis type 1 (NF1), extra-optic gliomas occur and may behave more aggressively with outcomes that differ by age. A retrospective case-control study was designed to describe the clinical course of adult NF1 patients with progressive extra-optic pilocytic astrocytomas (PAs) and compare to a pediatric cohort. Data for patients treated at the Johns Hopkins Comprehensive Neurofibromatosis Center from 2003 to 2013 were reviewed to identify cases (adults, age >18) and controls (pediatric, age <18) with clinically or radiographically progressive extra-optic PAs. Demographic, clinical, histologic, and radiographic data were collected. Three adult NF1 cases and four pediatric NF1 controls were identified. Mean age was 32.3 ± 9.5 years, 66% male (cases); 12.8 ± 4.2 years, 100% male (controls). Symptomatic progression occurred in two-of-three adults (67%) while the majority of pediatric patients presented with isolated radiographic progression (n = 3, 75%). Onset tended to be more rapid in adults (4 ± 1 vs. 14 ± 8.3 months, P = 0.10). Subtotal resection was the treatment for all pediatric patients. Radiotherapy (n = 2), chemotherapy (n = 2), and targeted, biologic agents (n = 2) were administered in adults. Although all pediatric patients are living, outcomes were universally poor in adults with progression to death in all (median survival 17.1 months, range 6.6-30.3). In conclusion, despite grade I histology, all three adult NF1 patients with progressive extra-optic PAs suffered an aggressive clinical course which was not seen in pediatric patients. Clinicians should be aware of this clinico-histologic discrepancy when counseling and managing adult NF1 patients with progressive extra-optic PAs. © 2016 Wiley Periodicals, Inc.


Subject(s)
Astrocytoma/diagnosis , Astrocytoma/etiology , Neurofibromatosis 1/complications , Adult , Astrocytoma/mortality , Astrocytoma/therapy , Biopsy , Brain/pathology , Case-Control Studies , Combined Modality Therapy , Diagnostic Imaging , Disease Progression , Female , Humans , Male , Neoplasm Grading , Neurofibromatosis 1/diagnosis , Phenotype , Retrospective Studies , Treatment Outcome , Young Adult
10.
Proc Natl Acad Sci U S A ; 110(44): 17933-8, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24114272

ABSTRACT

Glioblastoma (GBM), the most common brain malignancy, remains fatal with no effective treatment. Analyses of common aberrations in GBM suggest major regulatory pathways associated with disease etiology. However, 90% of GBMs are diagnosed at an advanced stage (primary GBMs), providing no access to early disease stages for assessing disease progression events. As such, both understanding of disease mechanisms and the development of biomarkers and therapeutics for effective disease management are limited. Here, we describe an adult-inducible astrocyte-specific system in genetically engineered mice that queries causation in disease evolution of regulatory networks perturbed in human GBM. Events yielding disease, both engineered and spontaneous, indicate ordered grade-specific perturbations that yield high-grade astrocytomas (anaplastic astrocytomas and GBMs). Impaired retinoblastoma protein RB tumor suppression yields grade II histopathology. Additional activation of v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) network drives progression to grade III disease, and further inactivation of phosphatase and tensin homolog (PTEN) yields GBM. Spontaneous missense mutation of tumor suppressor Trp53 arises subsequent to KRAS activation, but before grade III progression. The stochastic appearance of mutations identical to those observed in humans, particularly the same spectrum of p53 amino acid changes, supports the validity of engineered lesions and the ensuing interpretations of etiology. Absence of isocitrate dehydrogenase 1 (IDH1) mutation, asymptomatic low grade disease, and rapid emergence of GBM combined with a mesenchymal transcriptome signature reflect characteristics of primary GBM and provide insight into causal relationships.


Subject(s)
Astrocytoma/etiology , Biological Evolution , Disease Models, Animal , Genetic Engineering/methods , Glioblastoma/etiology , Animals , Base Sequence , Disease Progression , Gene Expression Profiling , Gene Regulatory Networks/genetics , Mice , Mice, Transgenic , Microarray Analysis , Molecular Sequence Data , Mutation, Missense/genetics , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Sequence Analysis, DNA , Tumor Suppressor Protein p53/genetics
11.
No To Hattatsu ; 48(6): 439-42, 2016 Nov.
Article in Japanese | MEDLINE | ID: mdl-30010299

ABSTRACT

Tuberous sclerosis complex (TSC) is a genetic disease affecting many organ systems and showing different symptoms in each age group. We encountered a TSC patient with intractable epilepsy who had brain tumors suspected to be subependymal giant cell astrocytoma (SEGA). We used adrenocorticotropic hormone and ordinal antiepileptic drugs at first, but they showed limited effectiveness. After we tried several treatments for epilepsy, we started to use everolimus to prevent tumor growth. As a result, the development of the tumor stopped and the epileptic attack improved simultaneously. The frequency and duration of each epileptic spasm and seizure became milder, and the electroencephalogram findings also improved. The mental development had regressed when the epilepsy started, but it started to progress again after the epileptic attack disappeared. Everolimus may be used for treatment of intractable epilepsy with TSC in patients with a growing SEGA.


Subject(s)
Astrocytoma/etiology , Spasms, Infantile/etiology , Tuberous Sclerosis/complications , Antineoplastic Agents/therapeutic use , Astrocytoma/drug therapy , Electroencephalography , Everolimus/therapeutic use , Female , Humans , Infant , Spasms, Infantile/physiopathology
12.
J Inherit Metab Dis ; 38(2): 273-7, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25338511

ABSTRACT

Patients with L-2-hydroxyglutaric aciduria are at risk for developing cerebral neoplasms, particularly gliomas, as one of the optical isomers of the known oncometabolite, 2-hydroxyglutarate is produced in L-2-hydroxyglutaric aciduria. To illustrate the concept of sustained oncogenic potential in permanent exposure to L-2-hydroxyglutarate in brain tissue, we present the medical history of a patient with L-2-hydroxyglutaric aciduria who underwent surgery to remove a right temporal anaplastic astrocytoma and developed an anatomically distinct, but histopathologically similar, tumor in the left frontal region 40 months later. This is the first reported case of successive distinct gliomas in a patient with L-2-hydroxyglutaric aciduria. While this implies a significant, cumulative lifetime risk for cerebral neoplasms in patients with this rare organic aciduria, it also allows further insight into a unique mechanism of tumorigenesis in the brain.


Subject(s)
Astrocytoma/etiology , Brain Diseases, Metabolic, Inborn/complications , Brain Neoplasms/etiology , Neoplasms, Second Primary/etiology , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Astrocytoma/genetics , Astrocytoma/metabolism , Astrocytoma/pathology , Astrocytoma/surgery , Biopsy , Brain Diseases, Metabolic, Inborn/diagnosis , Brain Diseases, Metabolic, Inborn/genetics , Brain Diseases, Metabolic, Inborn/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/surgery , ErbB Receptors/genetics , Genetic Predisposition to Disease , Glutarates/metabolism , Humans , In Situ Hybridization, Fluorescence , Magnetic Resonance Imaging , Male , Mutation , Neoplasm Grading , Neoplasms, Second Primary/genetics , Neoplasms, Second Primary/metabolism , Neoplasms, Second Primary/pathology , Positron-Emission Tomography , Risk Factors , Time Factors , Treatment Outcome , Young Adult
13.
Pediatr Blood Cancer ; 62(6): 1084-6, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25585602

ABSTRACT

Noonan syndrome (NS; MIM 163950) is an autosomal dominant syndrome which is clinically diagnosed by the distinct facial features, short stature, cardiac anomalies and developmental delay. About 50% of cases are associated with gain of function mutations in PTPN11 gene which leads to activation of the RAS/mitogen-activated protein kinase signaling pathway. This is known to have a role in tumorigenesis. Despite this, only limited reports of solid tumors (Fryssira H, Leventopoulos G, Psoni S, et al. Tumor development in three patients with Noonan syndrome. Eur J Pediatr 2008;167:1025-1031; Schuettpelz LG, McDonald S, Whitesell K et al. Pilocytic astrocytoma in a child with Noonan syndrome. Pediatr Blood Cancer 2009;53:1147-1149; Sherman CB, Ali-Nazir A, Gonzales-Gomez I, et al. Primary mixed glioneuronal tumor of the central nervous system in a patient with Noonan syndrome. J Pediatr Hematol Oncol 2009;31:61-64; Sanford RA, Bowman R, Tomita T, et al. A 16 year old male with Noonan's syndrome develops progressive scoliosis and deteriorating gait. Pediatr Neurosurg 1999;30:47-52) and no prior reports of optic gliomas have been described in patients with NS. We present here a patient with NS with a PTPN11 mutation and an optic pathway pilomyxoid astrocytoma.


Subject(s)
Astrocytoma/etiology , Noonan Syndrome/complications , Optic Nerve Neoplasms/etiology , Adolescent , Humans , Male , Mutation , Noonan Syndrome/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics
14.
Pediatr Blood Cancer ; 62(10): 1754-60, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25929843

ABSTRACT

BACKGROUND: Tuberous sclerosis complex (TSC) is an autosomal dominant multisystem disease usually diagnosed in childhood. Subependymal giant cell astrocytomas (SEGA) are benign brain lesions occurring in up to 20% of patients with TSC. Treatment with mTOR inhibitors has been proven effective in inducing SEGA shrinkage, but discontinuation results in re-growth. Evidence suggests that mTOR inhibition seems a disease-modifying treatment for TSC beyond inducing SEGA shrinkage; however concerns remain regarding negative long-term effects. METHODS: Through this retrospective case series, an attempt was made to determine the minimal mTOR inhibitor dose needed to maintain radiological response of SEGA in six pediatric patients treated at The Hospital for Sick Children since 2007. This study reviews medication doses and blood levels as related to SEGA size on MRIs at 3-month intervals. Blood levels were monitored every 3 months and 2 weeks after dose adjustment. Total dose reduction by 25% was considered after SEGA shrinkage was maintained on two consecutive MRIs. RESULTS: All patients demonstrated SEGA shrinkage greater than 50% when treated with mTOR inhibition at therapeutic doses (4-5 mg/m(2)). When sirolimus doses were weaned in two patients by 50%, SEGAs regrew by 84% and 32%. In two patients, responses have been maintained with 30% decrease in sirolimus dose. One patient underwent SEGA resection and one remains on therapeutic dose. CONCLUSIONS: Therapeutic dose of mTOR inhibitor is effective in shrinking TSC-related SEGAs. Doses less than 2.5 mg/m(2) were insufficient to maintain response in this limited series.


Subject(s)
Antineoplastic Agents/administration & dosage , Astrocytoma/drug therapy , Astrocytoma/pathology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tuberous Sclerosis/complications , Adolescent , Age of Onset , Astrocytoma/etiology , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Humans , Infant , Male , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Retrospective Studies
15.
Eur J Pediatr ; 174(8): 1069-76, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25749928

ABSTRACT

UNLABELLED: Common variable immunodeficiency (CVID) is a heterogeneous primary immunodeficiency associated with an increased risk of malignancy in adulthood, with lymphoma as one of the major causes of death. The aim of this study is to describe those malignancies detected in our cohort of pediatric CVID patients. We reviewed the clinical and laboratory data and the treatments and their outcomes in all pediatric CVID patients from our institution that developed a neoplasia. Four malignancies were diagnosed in three out of 27 pediatric CVID patients. Three malignancies were non-Hodgkin lymphoma (NHL) of B cell origin (mean age at diagnosis: 8 years old), and the remaining was a low-grade astrocytoma. Among NHL, two were mucosa-associated lymphoid tissue (MALT) lymphomas and one was associated with Epstein-Barr virus infection. NHL developed before CVID diagnosis in two patients. CVID patients showed different clinical phenotypes and belonged to different groups according Euroclass and Pediatric classification criteria. CONCLUSIONS: Malignancies, especially lymphoma, may develop in pediatric CVID patients with no previous signs of lymphoid hyperplasia and even before CVID diagnosis. Consequently, strategies for cancer prevention and/or early diagnosis are required in pediatric CVID patients.


Subject(s)
Astrocytoma/diagnosis , Common Variable Immunodeficiency/complications , Lymphoma, Non-Hodgkin/diagnosis , Adolescent , Astrocytoma/etiology , Astrocytoma/immunology , Child , Common Variable Immunodeficiency/immunology , Early Diagnosis , Female , Humans , Incidence , Lymphoma, Non-Hodgkin/etiology , Lymphoma, Non-Hodgkin/immunology , Male , Phenotype
16.
Proc Natl Acad Sci U S A ; 109(18): 6963-8, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22505738

ABSTRACT

Astrocytes are the most abundant cell of the CNS and demonstrate contact inhibition in which a nonproliferative, nonmotile cellular state is achieved once stable intercellular contacts are formed between mature cells. Cellular injury disrupts these intercellular contacts, causing a loss of contact inhibition and the rapid initiation of healing. Dysregulation of the molecular pathways involved in this process is thought to lead to an aggressive cellular state associated with neoplasia. We investigated whether a comparable correlation exists between the response of astrocytes to injury and the malignant phenotype of astrocytomas. We discovered that the loss of contact inhibition plays a critical role in the initiation and regulation of reactive astrocytes in the healing of wounds. In particular, injury of the astrocytes interrupts and destabilizes the cadherin-catenin complexes at the cell membrane leading to nuclear translocation of ß-catenin and characteristic changes associated with the activation of astrocytes. Similar signaling pathways are found to be active--but dysregulated--in astrocytomas. Inhibition of ß-catenin signaling diminished both the response of astrocytes to injury and induction of the malignant phenotype of astrocytomas. The findings shed light on a unique mechanism associated with the pathogenesis of astrocytomas and provide a model for the loss of contact inhibition that may broadly apply to understanding the mechanisms of tissue repair and tumorigenesis in the brain.


Subject(s)
Astrocytes/metabolism , Astrocytoma/etiology , Astrocytoma/metabolism , beta Catenin/metabolism , Animals , Astrocytoma/pathology , Cell Proliferation , Cell Transformation, Neoplastic , Cells, Cultured , Gene Knockdown Techniques , Mice , Models, Neurological , Phenotype , RNA, Small Interfering , Signal Transduction , Tumor Cells, Cultured , beta Catenin/antagonists & inhibitors , beta Catenin/genetics
17.
Przegl Lek ; 72(8): 435-9, 2015.
Article in Polish | MEDLINE | ID: mdl-26827561

ABSTRACT

Brain tumors in children are rare compared with other diseases childhood. Virtually every pediatrician working at the local hospital has been in contact or brain tumor diagnosis was that more half of them recognized the posterior fossa tumor. Analysis of surveys showed difficulties in the interpretation of basic neurological symptoms. Lessons learned from these studies point to the fact that the success of the surgery affects mainly the histological type of tumor and its location. It is extremely important to analyze complaints and symptoms young patients, we should listen to the parents of children with, because usually just parents are the first to notice that their offspring something was wrong.


Subject(s)
Astrocytoma/epidemiology , Infratentorial Neoplasms/epidemiology , Astrocytoma/etiology , Child , Cranial Fossa, Posterior , Female , Humans , Infratentorial Neoplasms/diagnosis , Infratentorial Neoplasms/etiology , Male , Poland/epidemiology
18.
J Pediatr ; 164(5): 1195-200, 2014 May.
Article in English | MEDLINE | ID: mdl-24518170

ABSTRACT

OBJECTIVES: To evaluate the efficacy and side effects of oral mammalian target of rapamycin (mTOR) inhibitors in children and adolescents with tuberous sclerosis complex (TSC) and intractable epilepsy or subependymal giant cell astrocytoma (SEGA). STUDY DESIGN: Single-center series of 13 children and adolescents with TSC who received sirolimus or everolimus (mTOR inhibitors). The anticonvulsant response was evaluated in 7 patients with TSC and refractory seizures. Six patients with SEGAs were treated with either sirolimus or everolimus for nonsurgical management. SEGA volumes were assessed longitudinally using 1.5-T magnetic resonance imaging. RESULTS: Of the intractable seizure group (7 patients), 1 patient had >90% reduction, 4 had 50%-90% reduction, and 2 had <50% reduction. Three reported subjective improvements in learning. By 12 months of treatment, there were statistically significant reductions in the SEGA volumes in 4 patients who received mTOR inhibitors (P < .04). The mean SEGA volume after 6 months of treatment was 2.18 cm(3), which represents 33% reduction in the mean baseline volume of 3.26 cm(3). The mTOR inhibitors were well tolerated. Adverse effects include dyslipidaemia (3 of 13), gingivitis (1 of 13), anorexia (1 of 13), and mild gastrointestinal side effects (1 of 13). CONCLUSION: This case series suggests that mTOR inhibitors can improve seizures in those with TSC and refractory epilepsy. They are also an effective treatment for reducing the volume of SEGAs in patients with TSC not amenable to surgery with an acceptable side effect profile.


Subject(s)
Anticonvulsants/therapeutic use , Antineoplastic Agents/therapeutic use , Astrocytoma/drug therapy , Epilepsy/drug therapy , Sirolimus/analogs & derivatives , Sirolimus/therapeutic use , Tuberous Sclerosis/complications , Adolescent , Astrocytoma/etiology , Child , Child, Preschool , Epilepsy/etiology , Everolimus , Female , Humans , Male , Prospective Studies , TOR Serine-Threonine Kinases/antagonists & inhibitors , Treatment Outcome
19.
Nephrol Dial Transplant ; 29(6): 1203-10, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24729041

ABSTRACT

BACKGROUND: Tuberous sclerosis complex (TSC) is characterized by benign tumours in multiple organs, including the brain, kidneys, skin, lungs and heart. Our objective was to evaluate everolimus, an mTOR inhibitor, in the treatment of angiomyolipoma in patients with subependymal giant cell astrocytoma (SEGA) associated with TSC. METHODS: EXamining everolimus In a Study of Tuberous Sclerosis Complex-1 (NCT00789828), a prospective, double-blind, randomized, placebo-controlled, Phase 3 study, examined everolimus in treating SEGA associated with TSC. Patients with serial SEGA growth from pre-baseline to baseline scans were randomly assigned (2:1) to receive 4.5 mg/m(2)/day everolimus (target blood trough: 5-15 ng/mL; n = 78) or placebo (n = 39). Angiomyolipoma response rates were analysed in patients (n = 44) with target baseline angiomyolipoma lesions (≥1 angiomyolipoma; longest diameter ≥1.0 cm). An angiomyolipoma response rate, defined as the proportion of patients with confirmed angiomyolipoma response, was assessed by kidney CT or MRI screening at baseline, at 12, 24 and 48 weeks and annually. RESULTS: Angiomyolipoma response rates were 53.3% (16/30) and 0% (0/14) for everolimus- and placebo-treated patients, respectively. Angiomyolipoma reductions ≥50% in the sum of volumes of all target lesions were seen only in everolimus-treated patients (56.5, 78.3 and 80.0%) compared with placebo-treated patients (0% at each time point) at Weeks 12, 24 and 48, respectively. Greater percentages of everolimus-treated patients had angiomyolipoma reductions ≥30% at these same time points (82.6, 100 and 100% versus 8.3, 18.2 and 16.7% for everolimus versus placebo, respectively). CONCLUSIONS: Everolimus showed efficacy in reducing angiomyolipoma lesion volume in patients with SEGA associated with TSC.The trial is registered with ClinicalTrials.gov, number NCT00789828; http://clinicaltrials.gov/ct2/show/NCT00789828?term=EXIST-1&rank=1.


Subject(s)
Angiomyolipoma/drug therapy , Antineoplastic Agents/therapeutic use , Astrocytoma/etiology , Kidney Neoplasms/drug therapy , Neoplasms, Multiple Primary/drug therapy , Sirolimus/analogs & derivatives , Tuberous Sclerosis/complications , Adult , Astrocytoma/epidemiology , Brain , Child, Preschool , Double-Blind Method , Everolimus , Female , Humans , Kidney Neoplasms/complications , Male , Prospective Studies , Protein Kinase Inhibitors/therapeutic use , Sirolimus/therapeutic use , Treatment Outcome , Tuberous Sclerosis/epidemiology
20.
Harefuah ; 153(1): 43-7, 63, 2014 Jan.
Article in Hebrew | MEDLINE | ID: mdl-24605407

ABSTRACT

Tuberous sclerosis complex (TSC) is a genetic syndrome, characterized by seizures, mental retardation, and a typical facial rash. Many patients have early onset seizures (usually during infancy) which are refractory to medical treatment. It has been shown that the neurocognitive decline in TSC patients is associated with the severity of seizures. The cerebral pathologies in TSC include - tubers (associated with seizures), benign tumors (subependymal giant cell astrocytomas - SEGAs), and intraventricular subependymal nodules (SENs). Until recent years, TSC patients with refractory seizures were only rarely considered to be suitable for epilepsy surgery. Over the past few years, several pre-surgical and surgical techniques have enabled patients to undergo epilepsy surgery with a high rate of seizure control. These techniques include invasive monitoring, mapping and resection of epileptogenic foci. The overall seizure outcome is around 75% seizure freedom. Additionally, benign brain tumors unique for TSC (SEGAs) present a treatment challenge. These tumors are typically located adjacent to the foramen of Monro. Without treatment the tumors may obstruct the CSF pathways leading to hydrocephalus. Current treatment options include surgical approaches, as well as medical treatment with mTOR inhibitors. The combination of medical and surgical treatments may decrease morbidity especially in partially resected tumors, multifocal tumors, or as neoadjuvant treatment in selected cases. In the current paper we review the neurosurgical aspects in the treatment of patients with TSC.


Subject(s)
Brain Neoplasms/surgery , Neurosurgical Procedures/methods , Tuberous Sclerosis/surgery , Astrocytoma/etiology , Astrocytoma/surgery , Brain Mapping/methods , Brain Neoplasms/etiology , Child , Combined Modality Therapy , Humans , Seizures/etiology , Seizures/surgery , Severity of Illness Index , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tuberous Sclerosis/physiopathology , Tuberous Sclerosis/therapy
SELECTION OF CITATIONS
SEARCH DETAIL