Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Invest Ophthalmol Vis Sci ; 65(6): 8, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38837168

ABSTRACT

Optic pathway gliomas (OPGs) are most predominant pilocytic astrocytomas, which are typically diagnosed within the first decade of life. The majority of affected children with OPGs also present with neurofibromatosis type 1 (NF1), the most common tumor predisposition syndrome. OPGs in individuals with NF1 primarily affect the optic pathway and lead to visual disturbance. However, it is challenging to assess risk in asymptomatic patients without valid biomarkers. On the other hand, for symptomatic patients, there is still no effective treatment to prevent or recover vision loss. Therefore, this review summarizes current knowledge regarding the pathogenesis of NF1-associated OPGs (NF1-OPGs) from preclinical studies to seek potential prognostic markers and therapeutic targets. First, the loss of the NF1 gene activates 3 distinct Ras effector pathways, including the PI3K/AKT/mTOR pathway, the MEK/ERK pathway, and the cAMP pathway, which mediate glioma tumorigenesis. Meanwhile, non-neoplastic cells from the tumor microenvironment (microglia, T cells, neurons, etc.) also contribute to gliomagenesis via various soluble factors. Subsequently, we investigated potential genetic risk factors, molecularly targeted therapies, and neuroprotective strategies for tumor prevention and vision recovery. Last, potential directions and promising preclinical models of NF1-OPGs are presented for further research. On the whole, NF1-OPGs develop as a result of the interaction between glioma cells and the tumor microenvironment. Developing effective treatments require a better understanding of tumor molecular characteristics, as well as multistage interventions targeting both neoplastic cells and non-neoplastic cells.


Subject(s)
Neurofibromatosis 1 , Optic Nerve Glioma , Humans , Neurofibromatosis 1/complications , Neurofibromatosis 1/genetics , Optic Nerve Glioma/therapy , Optic Nerve Glioma/genetics , Risk Factors , Animals , Neurofibromin 1/genetics , Optic Nerve Neoplasms/therapy , Optic Nerve Neoplasms/genetics
2.
Brain Tumor Pathol ; 38(1): 59-63, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33098465

ABSTRACT

Optic nerve glioma (ONG) is a rare, typically slow-growing WHO I grade tumor that affects the visual pathways. ONG is most commonly seen in the pediatric population, in association with neurofibromatosis type 1 syndrome. However, sporadic adult cases may also occur and may clinically behave more aggressively, despite benign histopathology. Genetic characterization of these tumors, particularly in the adult population, is lacking. A 39-year-old female presented with 1 month of progressive left-sided visual loss secondary to a enhancing mass along the left optic nerve sheath. Initial empiric management with focal radiotherapy failed to prevent tumor progression, prompting open biopsy which revealed a WHO I pilocytic astrocytoma of the optic nerve. Whole-exome sequencing of the biopsy specimen revealed somatic mutations in NF1,FGFR1 and PTPN11 that may provide actionable targets for molecularly guided therapies. Genetic characterization of ONG is lacking but is needed to guide the management of these rare but complex tumors. The genomic alterations reported in this case contributes to understanding the pathophysiology of adult sporadic ONG and may help guide future clinical prognostication and development of targeted therapies.


Subject(s)
Astrocytoma/genetics , Optic Nerve Glioma/genetics , Optic Nerve Neoplasms/genetics , Adult , Astrocytoma/pathology , Disease Progression , Female , Humans , Mutation , Neurofibromin 1/genetics , Optic Nerve Glioma/pathology , Optic Nerve Neoplasms/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Exome Sequencing
5.
J BUON ; 24(5): 2168-2172, 2019.
Article in English | MEDLINE | ID: mdl-31786891

ABSTRACT

PURPOSE: Farnesol has been shown to exhibit important anticancer potential. However, its antiproliferative effects have not been examined against the optic nerve sheath meningioma cells. In this study the potential of Farnesol in the treatment of optic nerve meningioma was evaluated by examining its antiproliferative effects against the HBL-52 cells. METHODS: The MTT assay was used to determine cell viability of HBL-52 cells. Autophagy was detected by transfection assay. The cell migration and invasion of HBL-52 cells was determined by transwell assay. Protein expression was checked by western blot assay. RESULTS: The results showed that Farnesol decreased significantly the viability of HBL-52 cells and showed an IC50 of 25 µM. The antiproliferative effects were due to the activation of the autophagy in the HBL-52 cells. The autophagy was also accompanied by upsurge of LC3 II and Beclin 1 expression. Farnesol also triggered the cell cycle arrest of the HBL-52 at the G2/M phase of the cell cycle which was accompanied by suppression of cyclin B1. The cell migration and invasion of the HBL-52 cells was also suppressed by Farnesol via inhibition of MMP-2 and 9 expressions. CONCLUSIONS: To sum up, Farnesol may prove beneficial in the treatment of optic nerve sheath meningioma as it has shown significant antiproliferative effects against this rare form of tumor.


Subject(s)
Cell Proliferation/drug effects , Farnesol/pharmacology , Meningioma/drug therapy , Optic Nerve Neoplasms/drug therapy , Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Meningioma/genetics , Meningioma/pathology , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Optic Nerve/drug effects , Optic Nerve/pathology , Optic Nerve Neoplasms/genetics , Optic Nerve Neoplasms/pathology , Signal Transduction/drug effects
7.
Pediatr Blood Cancer ; 65(3)2018 03.
Article in English | MEDLINE | ID: mdl-29049847

ABSTRACT

Children with neurofibromatosis type 1 (NF1) are predisposed to develop central nervous system neoplasms, the most common of which are low-grade gliomas (LGGs). The absence of human NF1 associated LGG-derived cell lines, coupled with an inability to generate patient-derived xenograft models, represents barriers to profile molecularly targeted therapies for these tumors. Thus, genetically engineered mouse models have been identified to evaluate the interplay between Nf1-deficient tumor cells and nonneoplastic stromal cells to evaluate potential therapies for these neoplasms. Future treatments might also consider targeting the nonneoplastic cells in NF1-LGGs to reduce tumor growth and neurologic morbidity in affected children.


Subject(s)
Glioma , Neoplasms, Experimental , Neurofibromatosis 1 , Neurofibromin 1/deficiency , Optic Nerve Neoplasms , Adolescent , Animals , Cell Line, Tumor , Child , Child, Preschool , Female , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Glioma/therapy , Humans , Infant , Infant, Newborn , Male , Mice , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Neurofibromatosis 1/genetics , Neurofibromatosis 1/metabolism , Neurofibromatosis 1/pathology , Neurofibromatosis 1/therapy , Optic Nerve Neoplasms/genetics , Optic Nerve Neoplasms/metabolism , Optic Nerve Neoplasms/pathology , Optic Nerve Neoplasms/therapy , Xenograft Model Antitumor Assays/methods
8.
Pediatr Hematol Oncol ; 34(4): 254-259, 2017 May.
Article in English | MEDLINE | ID: mdl-29040023

ABSTRACT

While clinical and radiographic responses to agents targeting the mitogen-activated protein kinases (MAPK) pathway have been repor-ted in pediatric low-grade gliomas (LGG), early phase trials indicate refractoriness to these medications in some of these patients. We report a patient with disseminated LGG with the BRAFV600E mutation, which was refractory to selumetinib, a MEK inhibitor, but subsequently showed immediate clinical and radiographic response to dabrafenib, a BRAF inhibitor, with sustained effect for 9 months prior to clinical progression. In LGGs, treatment resistance to one agent targeting the MAPK pathway might not imply refractoriness to other agents targeting this pathway.


Subject(s)
Benzimidazoles/administration & dosage , Chemoradiotherapy , Glioma , Imidazoles/administration & dosage , Mutation, Missense , Optic Nerve Neoplasms , Oximes/administration & dosage , Proto-Oncogene Proteins B-raf , Amino Acid Substitution , Glioma/enzymology , Glioma/genetics , Glioma/pathology , Glioma/therapy , Humans , Infant , Male , Optic Nerve Neoplasms/enzymology , Optic Nerve Neoplasms/genetics , Optic Nerve Neoplasms/pathology , Optic Nerve Neoplasms/therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism
9.
J Neuroophthalmol ; 36(1): 73-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26628337

ABSTRACT

Bilateral, simultaneous optic nerve sheath infiltration as a manifestation of leukemia relapse is very rare. A 45-year-old woman with chronic myelogenous leukemia was successfully treated to cytogenetic bone marrow remission 1 year previously and maintained on imatinib. She developed total bilateral blindness with marked, bilateral optic disc edema and evidence of bilateral optic nerve infiltration on magnetic resonance imaging. Cerebrospinal fluid cytology confirmed central nervous system (CNS) blast crisis. She recovered visual acuity of 20/20 in the right eye, and 20/25 in the left eye with salvage systemic and intrathecal chemotherapy before radiation therapy. Our report underscores the importance of timely and aggressive intervention of blast crisis of the CNS and the need for CNS penetrating induction and maintenance therapy.


Subject(s)
Blast Crisis/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemic Infiltration/pathology , Neoplasm Recurrence, Local/pathology , Optic Nerve Neoplasms/pathology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blast Crisis/drug therapy , Blast Crisis/genetics , Female , Genes, abl/genetics , Humans , In Situ Hybridization, Fluorescence , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemic Infiltration/drug therapy , Magnetic Resonance Imaging , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Optic Nerve Neoplasms/drug therapy , Optic Nerve Neoplasms/genetics , Real-Time Polymerase Chain Reaction , Salvage Therapy , Tomography, Optical Coherence
10.
Middle East Afr J Ophthalmol ; 22(1): 117-8, 2015.
Article in English | MEDLINE | ID: mdl-25624686

ABSTRACT

We present a case of a 3-year-old girl with a positive family history of neurofibromatosis type-1 (NF1) presented with best corrected visual acuity of 20/40 in the right eye and <20/400 in the left eye. External ocular examination revealed left eye proptosis of 3 mm, grade II left relative afferent pupillary defect and full range of ocular motility with no strabismus. Slit lamp examination revealed iris lisch nodules bilaterally. Dilated fundus examination of the right eye was normal. Left eye disclosed a large mass extending from the optic nerve head, with associated subretinal fluid. There was neovascularization at the optic disc as well as a superior retinal hemorrhage. Computed tomography of brain/orbits showed an enlarged left optic nerve with a large mass at the optic nerve head, with no evidence of calcification. In addition, a large left optic pathway glioma (OPG), multiple hamartomas within the brain and a smaller low-grade right OPG was also reported. The remarkable feature of our case is the rare intraocular optic nerve involvement of the OPG. Early and regular ophthalmological assessment of all NF1 suspect/confirmed cases is of paramount importance in order to detect OPG early, resulting in timely intervention and salvage of vision.


Subject(s)
Neurofibromatosis 1/pathology , Optic Nerve Glioma/pathology , Optic Nerve Neoplasms/pathology , Antineoplastic Agents, Phytogenic/therapeutic use , Child, Preschool , Female , Humans , Magnetic Resonance Imaging , Neurofibromatosis 1/drug therapy , Neurofibromatosis 1/genetics , Neurofibromin 1/genetics , Optic Disk/pathology , Optic Nerve Glioma/drug therapy , Optic Nerve Glioma/genetics , Optic Nerve Neoplasms/drug therapy , Optic Nerve Neoplasms/genetics , Point Mutation , Subretinal Fluid , Vinblastine/therapeutic use , Visual Acuity/physiology
11.
Neuro Oncol ; 17(5): 670-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25246427

ABSTRACT

BACKGROUND: Children with the neurofibromatosis type 1 (NF1) tumor predisposition syndrome are prone to the development of optic pathway gliomas resulting from biallelic inactivation of the NF1 gene. Recent studies have revealed the presence of other molecular alterations in a small portion of these NF1-associated brain tumors. The purpose of this study was to leverage Nf1 genetically engineered mouse strains to define the functional significance of these changes to optic glioma biology. METHODS: Nf1+/- mice were intercrossed with Nf1(flox/flox) mice, which were then crossed with Nf1(flox/flox); GFAP-Cre mice, to generate Nf1(flox/mut); GFAP-Cre (FMC) mice. These mice were additionally mated with conditional KIAA1549:BRAF knock-in or Pten(flox/wt) mice to generate Nf1(flox/mut); f-BRAF; GFAP-Cre (FMBC) mice or Nf1(flox/mut); Pten(flox/wt); GFAP-Cre (FMPC) mice, respectively. The resulting optic gliomas were analyzed for changes in tumor volume, proliferation, and retinal ganglion cell loss. RESULTS: While KIAA1549:BRAF conferred no additional biological properties on Nf1 optic glioma, FMPC mice had larger optic gliomas with greater proliferative indices and microglial infiltration. In addition, all 3 Nf1 murine optic glioma strains exhibited reduced retinal ganglion cell survival and numbers; however, FMPC mice had greater retinal nerve fiber layer thinning near the optic head relative to FMC and FMBC mice. CONCLUSIONS: Collectively, these experiments demonstrate genetic cooperativity between Nf1 loss and Pten heterozygosity relevant to optic glioma biology and further underscore the value of employing genetically engineered mouse strains to define the contribution of discovered molecular alterations to brain tumor pathogenesis.


Subject(s)
Neurofibromatosis 1/genetics , Optic Nerve Glioma/genetics , Optic Nerve Neoplasms/genetics , Animals , Cell Death , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Optic Nerve Glioma/metabolism , Optic Nerve Glioma/pathology , Optic Nerve Neoplasms/metabolism , Optic Nerve Neoplasms/pathology , PTEN Phosphohydrolase/genetics , Retinal Ganglion Cells/pathology , Signal Transduction
12.
Brain Pathol ; 25(2): 193-201, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24773246

ABSTRACT

Orbital meningiomas can be classified as primary optic nerve sheath (ON) meningiomas, primary intraorbital ectopic (Ob) meningiomas and spheno-orbital (Sph-Ob) meningiomas based on anatomic site. Single-nucleotide polymorphism (SNP)-based array analysis with the Illumina 300K platform was performed on formalin-fixed, paraffin-embedded tissue from 19 orbital meningiomas (5 ON, 4 Ob and 10 Sph-Ob meningiomas). Tumors were World Health Organization (WHO) grade I except for two grade II meningiomas, and one was NF2-associated. We found genomic alterations in 68% (13 of 19) of orbital meningiomas. Sph-Ob tumors frequently exhibited monosomy 22/22q loss (70%; 7/10) and deletion of chromosome 1p, 6q and 19p (50% each; 5/10). Among genetic alterations, loss of chromosome 1p and 6q were more frequent in clinically progressive tumors. Chromosome 22q loss also was detected in the majority of Ob meningiomas (75%; 3/4) but was infrequent in ON meningiomas (20%; 1/5). In general, Ob tumors had fewer chromosome alterations than Sph-Ob and ON tumors. Unlike Sph-Ob meningiomas, most of the Ob and ON meningiomas did not progress even after incomplete excision, although follow-up was limited in some cases. Our study suggests that ON, Ob and Sph-Ob meningiomas are three molecularly distinct entities. Our results also suggest that molecular subclassification may have prognostic implications.


Subject(s)
Meningioma/genetics , Optic Nerve Neoplasms/genetics , Orbital Neoplasms/genetics , Polymorphism, Single Nucleotide , Adult , Aged , Chromosome Deletion , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 22/genetics , Chromosomes, Human, Pair 6/genetics , Female , Humans , Male , Meningioma/classification , Middle Aged , Optic Nerve Neoplasms/classification , Orbital Neoplasms/classification , Young Adult
13.
Ophthalmic Plast Reconstr Surg ; 30(5): 372-6, 2014.
Article in English | MEDLINE | ID: mdl-24762952

ABSTRACT

PURPOSE: This study was designed to better understand the biologic nature of optic nerve gliomas (ONGs) and to investigate staining techniques that might improve the pathologic interpretation of surgical margins. METHODS: In this retrospective case series, clinical data on patient presentation, MRI, surgical visualization, and initial pathologic interpretation were gathered. Specimens were then reexamined using analysis of p53, isocitrate dehydrogenase 1 (IDH1), MIB-1, and B-rapidly accelerated fibrosarcoma (BRAF) duplication. RESULTS: Six patients were studied. All were diagnosed with World Health Organization grade 1 ONGs on original pathology. On reexamination, BRAF tandem duplication was found in 2 patients with neurofibromatosis Type 1 association. P53 immunoreactivity was noted in a third case. No cases had IDH1 immunoreactivity. Focal elevations of MIB-1 up to 7.5% were noted in 2 cases. CONCLUSIONS: ONGs are neoplasms with variable degrees of aggressiveness. As more is understood regarding their varied genetic underpinnings, improved pathologic classification and individualized treatment regimens may be achieved. The authors hope that this study helps guide the oculoplastic community toward a multi-institutional, prospective study of ONG genomic sequencing.


Subject(s)
Optic Nerve Glioma , Optic Nerve Neoplasms , Adolescent , Biomarkers, Tumor/metabolism , Child , Child, Preschool , Female , Humans , In Situ Hybridization, Fluorescence , Isocitrate Dehydrogenase/metabolism , Ki-67 Antigen/metabolism , Magnetic Resonance Imaging , Male , Optic Nerve Glioma/genetics , Optic Nerve Glioma/metabolism , Optic Nerve Glioma/pathology , Optic Nerve Neoplasms/genetics , Optic Nerve Neoplasms/metabolism , Optic Nerve Neoplasms/pathology , Proto-Oncogene Proteins B-raf/metabolism , Retrospective Studies , Tumor Suppressor Protein p53/metabolism
14.
Acta Neuropathol ; 127(4): 573-91, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24232507

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) are Schwann cell-derived malignancies that arise from plexiform neurofibromas in patients with mutation of the neurofibromin 1 (NF1) gene. We have shown that the growth factor neuregulin-1 (NRG1) also contributes to human neurofibroma and MPNST pathogenesis and that outbred C57BL/6J × SJL/J transgenic mice overexpressing NRG1 in Schwann cells (P0-GGFß3 mice) recapitulate the process of neurofibroma-MPNST progression. However, it is unclear whether NRG1 acts predominantly within NF1-regulated signaling cascades or instead activates other essential cascades that cooperate with NF1 loss to promote tumorigenesis. We now report that tumorigenesis is suppressed in inbred P0-GGFß3 mice on a C57BL/6J background. To determine whether NRG1 overexpression interacts with reduced Nf1 or Trp53 gene dosage to "unmask" tumorigenesis in these animals, we followed cohorts of inbred P0-GGFß3;Nf1+/−, P0-GGFß3;Trp53+/− and control (P0-GGFß3, Nf1+/− and Trp53+/−) mice for 1 year. We found no reduction in survival or tumors in control and P0-GGFß3;Nf1+/− mice. In contrast, P0-GGFß3;Trp53+/− mice died on average at 226 days, with MPNSTs present in 95 % of these mice. MPNSTs in inbred P0-GGFß3;Trp53+/− mice arose de novo from micro-MPNSTs that uniformly develop intraganglionically. These micro-MPNSTs are of lower grade (WHO grade II-III) than the major MPNSTs (WHO grade III-IV); array comparative genomic hybridization showed that lower grade MPNSTs also had fewer genomic abnormalities. Thus, P0-GGFß3;Trp53+/− mice represent a novel model of low- to high-grade MPNST progression. We further conclude that NRG1 promotes peripheral nervous system neoplasia predominantly via its effects on the signaling cascades affected by Nf1 loss.


Subject(s)
Gene Expression , Haploinsufficiency/genetics , Neuregulin-1/metabolism , Optic Nerve Neoplasms/genetics , Optic Nerve Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Animals , Desmin/metabolism , Humans , In Situ Nick-End Labeling , Ki-67 Antigen/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Myelin P0 Protein/genetics , Myelin P0 Protein/metabolism , Neurofibroma/genetics , S100 Calcium Binding Protein beta Subunit/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Nerve Roots/metabolism , Spinal Nerve Roots/pathology
15.
Mod Pathol ; 26(10): 1279-87, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23702730

ABSTRACT

Pilocytic astrocytoma is a low-grade glioma that affects mostly children and young adults and can occur anywhere in the central nervous system. Pilocytic astrocytoma of the optic nerve is an equally indolent subtype that is occasionally associated with neurofibromatosis type 1. In earlier studies, this subtype was considered within the larger category of 'optic pathway glioma,' which included infiltrating astrocytomas and other hypothalamic tumors. However, there have been suggestions that gliomas in the optic nerve, and especially pilocytic astrocytoma of the optic nerve, are biologically different from tumors within the hypothalamus and other parts of the optic tract. Furthermore, the recent discovery of BRAF duplication and fusion with the KIAA1549 gene is reported to be more typical for posterior fossa tumors, and the rate of this aberration is not well known in pilocytic astrocytoma of the optic nerve. To determine the distinction of pilocytic astrocytoma of the optic nerve from pilocytic astrocytoma of the posterior fossa and to investigate the prevalence of BRAF aberrations, we reviewed the clinicopathological and molecular features of all such patients in our institution. Our study demonstrates that BRAF duplication is more frequent in posterior fossa tumors compared with pilocytic astrocytoma of the optic nerve (P=0.011). However, the rates of phospho-MAPK1 and CDKN2A expression were high in both pilocytic astrocytoma of the optic nerve and posterior fossa pilocytic astrocytoma, suggesting that the MAPK pathway is active in these tumors. Our study supports the notion that BRAF duplication is more typical of posterior fossa pilocytic astrocytoma and that molecular alterations other than KIAA1549 fusion may underlie MAPK pathway activation in pilocytic astrocytoma of the optic nerve.


Subject(s)
Astrocytoma/pathology , Central Nervous System Neoplasms/pathology , Optic Nerve Neoplasms/pathology , Proto-Oncogene Proteins B-raf/genetics , Astrocytoma/genetics , Astrocytoma/metabolism , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/metabolism , Child , Child, Preschool , DNA Mutational Analysis , Female , Humans , Infant , Male , Optic Nerve Neoplasms/genetics , Optic Nerve Neoplasms/metabolism , Proto-Oncogene Proteins B-raf/metabolism
16.
J Child Neurol ; 28(5): 625-32, 2013 May.
Article in English | MEDLINE | ID: mdl-23439714

ABSTRACT

Pilocytic astrocytoma, the most common pediatric brain tumor, is a clinically and molecularly heterogeneous disease that occurs most often in the cerebellum and hypothalamic and chiasmatic regions. Classically, pilocytic astrocytomas are driven by the mitogen-activated protein kinase/extracellular signal-regulated kinase pathway. Recently described genetic aberrations involving this pathway are critical for tumorigenesis. Tandem duplication of 7q34 encodes BRAF and produces several KIAA1549-BRAF novel oncogenic fusions. Activating point mutations of BRAF, such as BRAF (V600E), also lead to pilocytic astrocytoma. Loss of the NF1 gene allows hyperactivation of the oncogene KRAS. In this review, we discuss the current understanding of the novel molecular aberrations described in pilocytic astrocytomas and their clinical relevance for prognosis and treatment. The prognostic indications of these aberrations are discussed with regard to tumor location, tumor pathology, and patient age. A better understanding of the evolving molecular heterogeneity of pilocytic astrocytomas offers hope for developing molecularly targeted therapeutic armamentariums.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Chromosome Aberrations , Genetic Heterogeneity , Astrocytoma/diagnosis , Astrocytoma/therapy , Brain Neoplasms/diagnosis , Brain Neoplasms/therapy , Cell Transformation, Neoplastic/genetics , Cerebellar Neoplasms/diagnosis , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/therapy , Child , Extracellular Signal-Regulated MAP Kinases/genetics , Genes, Neurofibromatosis 1 , Humans , Hypothalamic Neoplasms/diagnosis , Hypothalamic Neoplasms/genetics , Hypothalamic Neoplasms/therapy , Molecular Targeted Therapy , Optic Chiasm , Optic Nerve Neoplasms/diagnosis , Optic Nerve Neoplasms/genetics , Optic Nerve Neoplasms/therapy , Point Mutation/genetics , Prognosis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras) , Transcriptional Activation/genetics , ras Proteins/genetics
17.
J Neurosurg ; 119(2): 494-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23432529

ABSTRACT

Meningiomas rarely cause CSF dissemination, and CSF seeding to the optic nerve (ON) is extremely rare. This is the first report of 2 cases of atypical meningioma with subacute visual loss due to ON seeding. The authors present the genetic characteristics of these atypical meningiomas with CSF dissemination. The patient in Case 1 was a 36-year-old woman with a 1.5-cm mass within the left ON, and the patient in Case 2 was a 70-year-old woman with a 0.9-cm mass around the right ON. Both individuals had undergone multiple surgeries for primary lesions and local recurrent lesions. They presented with subacute visual loss, and both tumors were completely resected. The pathological diagnosis was atypical meningioma with high MIB-1 indices and p53-positive cell ratios in each case. Comparative genomic hybridization showed significant chromosomal copy number alterations similar to the results of previous surgeries, confirming that the tumors were disseminated lesions. The present findings suggest that genetic characteristics, such as 1p and 10qcen-23 losses and 17q and 20 gains, shared by the 2 cases might be associated with CSF dissemination of meningiomas.


Subject(s)
Meningeal Neoplasms/pathology , Meningioma/pathology , Neoplasm Seeding , Optic Nerve Neoplasms/pathology , Optic Nerve/pathology , Vision, Low/etiology , Adult , Aged , DNA Copy Number Variations , Female , Humans , Meningeal Neoplasms/complications , Meningeal Neoplasms/genetics , Meningeal Neoplasms/surgery , Meningioma/complications , Meningioma/genetics , Meningioma/surgery , Optic Nerve Neoplasms/complications , Optic Nerve Neoplasms/genetics , Optic Nerve Neoplasms/surgery
19.
Brain ; 132(Pt 6): 1523-35, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19336457

ABSTRACT

Pilocytic astrocytomas are WHO grade I gliomas that occur predominantly in childhood. They share features of both astroglial and oligodendroglial lineages. These tumours affect preferentially the cerebellum (benign clinical course) and the optic pathway, especially the hypothalamo-chiasmatic region (poor prognosis). Understanding the molecular basis responsible for the aggressive behaviour of hypothalamo-chiasmatic pilocytic astrocytomas is a prerequisite to setting up new molecular targeted therapies. We used the microarray technique to compare the transcriptional profiles of five hypothalamo-chiasmatic and six cerebellar pilocytic astrocytomas. Validation of the microarray results and comparison of the tumours with normal developing tissue was done by quantitative real-time PCR and immunohistochemistry. Results demonstrate that cerebellar and hypothalamo-chiasmatic pilocytic astrocytomas are two genetically distinct and topography-dependent entities. Numerous genes upregulated in hypothalamo-chiasmatic pilocytic astrocytomas also increased in the developing chiasm, suggesting that developmental genes mirror the cell of origin whereas migrative, adhesive and proliferative genes reflect infiltrative properties of these tumours. Of particular interest, NOTCH2, a gene expressed in radial glia and involved in gliomagenesis, was upregulated in hypothalamo-chiasmatic pilocytic astrocytomas. In order to find progenitor cells that could give rise to hypothalamo-chiasmatic pilocytic astrocytomas, we performed a morphological study of the hypothalamo-chiasmatic region and identified, in the floor of the third ventricle, a unique population of vimentin- and glial fibrillary acidic protein-positive cells highly suggestive of radial glia cells. Therefore, pilocytic astrocytomas of the hypothalamo-chiasmatic region should be considered as a distinct entity which probably originates from a unique population of cells with radial glia phenotype.


Subject(s)
Astrocytoma/diagnosis , Optic Nerve Neoplasms/diagnosis , Adolescent , Adult , Astrocytes/metabolism , Astrocytoma/genetics , Astrocytoma/pathology , Cell Proliferation , Cerebellar Neoplasms/diagnosis , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Child , Child, Preschool , DNA, Neoplasm/genetics , Diagnosis, Differential , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Hypothalamus/metabolism , Infant , Middle Aged , Neoplastic Stem Cells/pathology , Neuroglia/pathology , Oligonucleotide Array Sequence Analysis/methods , Optic Chiasm/cytology , Optic Chiasm/embryology , Optic Chiasm/metabolism , Optic Nerve Neoplasms/genetics , Optic Nerve Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction/methods , Up-Regulation , Vimentin/metabolism , Young Adult
20.
Klin Oczna ; 111(10-12): 378-83, 2009.
Article in Polish | MEDLINE | ID: mdl-20169901

ABSTRACT

PURPOSE: This study was to evaluate pathogenesis, symptoms, clinical course and possible treatment of neurofibromatosis type 1. Neurofibromatosis type 1, von Recklinghausen's disease, is one of the phacomatoses. It belongs to the most frequent inherited diseases in human population. This disease is autosomal dominant, but new spontaneous mutation are also common. The symptoms are caused by disorders in the melanocytes and gliocytes. In ophthalmological examination Lisch nodules, café-au lait spots, neurofibromas of the lids, optic pathway gliomas and deformation of the orbit bones, can be observed. The symptoms are different, depend upon the age and demonstrate charateristic evolution through the lifetime.


Subject(s)
Eye Neoplasms/diagnosis , Neurofibromatosis 1/diagnosis , Optic Nerve Glioma/diagnosis , Conjunctival Neoplasms/diagnosis , Conjunctival Neoplasms/genetics , Eye Neoplasms/genetics , Humans , Neurofibromatosis 1/genetics , Optic Nerve Glioma/genetics , Optic Nerve Neoplasms/diagnosis , Optic Nerve Neoplasms/genetics , Skin Neoplasms/diagnosis , Skin Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...