151
|
de Blank P, Fisher MJ, Gittleman H, Barnholtz-Sloan JS, Badve C, Berman JI. Validation of an automated tractography method for the optic radiations as a biomarker of visual acuity in neurofibromatosis-associated optic pathway glioma. Exp Neurol 2017; 299:308-316. [PMID: 28587872 DOI: 10.1016/j.expneurol.2017.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 04/21/2017] [Accepted: 06/02/2017] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Fractional anisotropy (FA) of the optic radiations has been associated with vision deficit in multiple intrinsic brain pathologies including NF1 associated optic pathway glioma, but hand-drawn regions of interest used in previous tractography methods limit consistency of this potential biomarker. We created an automated method to identify white matter tracts in the optic radiations and compared this method to previously reported hand-drawn tractography. METHOD Automated tractography of the optic radiation using probabilistic streamline fiber tracking between the lateral geniculate nucleus of the thalamus and the occipital cortex was compared to the hand-drawn method between regions of interest posterior to Meyer's loop and anterior to tract branching near the calcarine cortex. Reliability was assessed by two independent raters in a sample of 20 healthy child controls. Among 50 children with NF1-associated optic pathway glioma, the association of FA and visual acuity deficit was compared for both tractography methods. RESULTS Hand-drawn tractography methods required 2.6±0.9min/participant; automated methods were performed in <1min of operator time for all participants. Cronbach's alpha was 0.83 between two independent raters for FA in hand-drawn tractography, but repeated automated tractography resulted in identical FA values (Cronbach's alpha=1). On univariate and multivariate analyses, FA was similarly associated with visual acuity loss using both methods. Receiver operator characteristic curves of both multivariate models demonstrated that both automated and hand-drawn tractography methods were equally able to distinguish normal from abnormal visual acuity. CONCLUSION Automated tractography of the optic radiations offers a fast, reliable and consistent method of tract identification that is not reliant on operator time or expertise. This method of tract identification may be useful as DTI is developed as a potential biomarker for visual acuity.
Collapse
Affiliation(s)
- Peter de Blank
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; University of Cincinnati, Department of Pediatrics, Cincinnati, OH, United States.
| | - Michael J Fisher
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Haley Gittleman
- University of Cincinnati, Department of Pediatrics, Cincinnati, OH, United States
| | | | - Chaitra Badve
- University of Cincinnati, Department of Pediatrics, Cincinnati, OH, United States
| | - Jeffrey I Berman
- Department of Radiology, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| |
Collapse
|
152
|
Trevisson E, Cassina M, Opocher E, Vicenzi V, Lucchetta M, Parrozzani R, Miglionico G, Mardari R, Viscardi E, Midena E, Clementi M. Natural history of optic pathway gliomas in a cohort of unselected patients affected by Neurofibromatosis 1. J Neurooncol 2017; 134:279-287. [PMID: 28577031 DOI: 10.1007/s11060-017-2517-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 05/29/2017] [Indexed: 11/28/2022]
Abstract
Optic pathway glioma (OPG) represents the most common central nervous system tumor in children with Neurofibromatosis type-1 (NF1). Although overall survival is usually good, no clear prognostic factors have been identified so far. We assessed the natural history of OPG in a cohort of unselected patients affected by NF1. We retrospectively evaluated 414 consecutive patients affected by NF1 and referred to our NF1 clinic before age 6. Average follow-up was 11.9 years: 52 out of 414 patients had OPG with a total cumulative incidence of 15.4% at age 15 (Kaplan-Meier estimate) and a statistically significant difference according to sex. Brain and orbit MRI was performed in 44.7% of patients: 34.6% for screening purposes and 65.4% because of the presence of neurological, ocular or other symptoms. OPG was diagnosed in 12.5% of cases in the first group, whereas in 36.4% in the latter group (p = 0.001). Clinical management was conservative in most patients, while 8 of them underwent therapy mainly because of visual deterioration. OPG was diagnosed earlier in treated patients, but the difference was not statistically significant. Conversely, all patients who underwent screening MRI had normal visual outcome. In conclusion, OPG location does not correlate with need for treatment; female patients were more frequently affected by OPG but not more frequently treated. OPG diagnosis by screening MRI does not affect the natural history of the tumor.
Collapse
Affiliation(s)
- Eva Trevisson
- Clinical Genetics Unit, Department of Woman and Child Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy.
| | - Matteo Cassina
- Clinical Genetics Unit, Department of Woman and Child Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy
| | - Enrico Opocher
- Hematology-Oncology Clinic, Department of Woman and Child Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy
| | - Virginia Vicenzi
- Clinical Genetics Unit, Department of Woman and Child Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy
| | - Marta Lucchetta
- Neurological Unit, Rovigo General Hospital, Viale Tre Martiri, 140, 45100, Rovigo, Italy
| | - Raffaele Parrozzani
- Department of Neurosciences, University of Padova, Via Giustiniani 2, 35128, Padova, Italy
| | - Giacomo Miglionico
- G.B. Bietti Eye Foundation, IRCCS, Ocular Oncology and Toxicology Research Unit, Via Livenza, 3, 00198, Roma, Italy
| | - Rodica Mardari
- Neuroradiology Unit, Hospital of Padova, Via Giustiniani 2, 35128, Padova, Italy
| | - Elisabetta Viscardi
- Hematology-Oncology Clinic, Department of Woman and Child Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy
| | - Edoardo Midena
- Department of Neurosciences, University of Padova, Via Giustiniani 2, 35128, Padova, Italy.,G.B. Bietti Eye Foundation, IRCCS, Ocular Oncology and Toxicology Research Unit, Via Livenza, 3, 00198, Roma, Italy
| | - Maurizio Clementi
- Clinical Genetics Unit, Department of Woman and Child Health, University of Padova, Via Giustiniani 3, 35128, Padova, Italy
| |
Collapse
|
153
|
Packer RJ, Pfister S, Bouffet E, Avery R, Bandopadhayay P, Bornhorst M, Bowers DC, Ellison D, Fangusaro J, Foreman N, Fouladi M, Gajjar A, Haas-Kogan D, Hawkins C, Ho CY, Hwang E, Jabado N, Kilburn LB, Lassaletta A, Ligon KL, Massimino M, Meeteren SV, Mueller S, Nicolaides T, Perilongo G, Tabori U, Vezina G, Warren K, Witt O, Zhu Y, Jones DT, Kieran M. Pediatric low-grade gliomas: implications of the biologic era. Neuro Oncol 2017; 19:750-761. [PMID: 27683733 PMCID: PMC5464436 DOI: 10.1093/neuonc/now209] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
For the past decade, it has been recognized that pediatric low-grade gliomas (LGGs) and glial-neuronal tumors carry distinct molecular alterations with resultant aberrant intracellular signaling in the Ras-mitogen-activated protein kinase pathway. The conclusions and recommendations of a consensus conference of how best to integrate the growing body of molecular genetic information into tumor classifications and, more importantly, for future treatment of pediatric LGGs are summarized here. There is uniform agreement that molecular characterization must be incorporated into classification and is increasingly critical for appropriate management. Molecular-targeted therapies should be integrated expeditiously, but also carefully into the management of these tumors and success measured not only by radiographic responses or stability, but also by functional outcomes. These trials need to be carried out with the caveat that the long-term impact of molecularly targeted therapy on the developing nervous system, especially with long duration treatment, is essentially unknown.
Collapse
Affiliation(s)
- Roger J Packer
- Center for Neuroscience and Behavioral Medicine, Washington, District of Columbia, USA
- Gilbert Family Neurofibromatosis Institute, Washington, District of Columbia, USA
- Brain Tumor Institute, Washington, District of Columbia, USA
| | - Stephan Pfister
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Eric Bouffet
- Paediatric Neuro-Oncology Program, Research Institute and The Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Robert Avery
- Center for Neuroscience and Behavioral Medicine, Washington, District of Columbia, USA
- Gilbert Family Neurofibromatosis Institute, Washington, District of Columbia, USA
- Brain Tumor Institute, Washington, District of Columbia, USA
| | - Pratiti Bandopadhayay
- Department of Pediatrics, Brigham and Women's Hospital, Harvard Medical School, and the Broad Institute, Dana-Farber/Boston Children's Cancer and Blood Disorders Centre, Boston, Massachusetts, USA
| | - Miriam Bornhorst
- Center for Neuroscience and Behavioral Medicine, Washington, District of Columbia, USA
- Gilbert Family Neurofibromatosis Institute, Washington, District of Columbia, USA
- Brain Tumor Institute, Washington, District of Columbia, USA
- Center for Cancer and Immunology Research, Washington, District of Columbia, USA
| | - Daniel C Bowers
- Department of Pediatrics, UT Southwestern Medical School, Dallas, Texas, USA
| | - David Ellison
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee. USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jason Fangusaro
- Ann and Robert H. Lurie Children's Hospital of Chicago Department of Pediatric Hematology/Oncology and Stem Cell Transplantation, University of Colorado, Aurora, Colorado, USA
| | - Nicholas Foreman
- Northwestern Feinberg School of Medicine, Chicago, Illinois; Children's Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| | - Maryam Fouladi
- Brain Tumor Center, Brain Tumor Translational Research, UC Department of Pediatrics, Cincinnati, Ohio, USA
| | - Amar Gajjar
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Daphne Haas-Kogan
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, and the Broad Institute, Dana-Farber/Boston Children's Cancer and Blood Disorders Centre, Boston, Massachusetts, USA
| | - Cynthia Hawkins
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee. USA
| | - Cheng-Ying Ho
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Eugene Hwang
- Center for Neuroscience and Behavioral Medicine, Washington, District of Columbia, USA
- Brain Tumor Institute, Washington, District of Columbia, USA
- Center for Cancer and Immunology Research, Washington, District of Columbia, USA
| | - Nada Jabado
- Ann and Robert H. Lurie Children's Hospital of Chicago Department of Pediatric Hematology/Oncology and Stem Cell Transplantation, University of Colorado, Aurora, Colorado, USA
| | - Lindsay B Kilburn
- Brain Tumor Institute, Washington, District of Columbia, USA
- Center for Cancer and Immunology Research, Washington, District of Columbia, USA
| | - Alvaro Lassaletta
- Northwestern Feinberg School of Medicine, Chicago, Illinois; Children's Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| | - Keith L Ligon
- Brain Tumor Center, Brain Tumor Translational Research, UC Department of Pediatrics, Cincinnati, Ohio, USA
- Brain Tumor Center, Brain Tumor Translational Research, UC Department of Pediatrics, Cincinnati, Ohio, USA
| | - Maura Massimino
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, and the Broad Institute, Dana-Farber/Boston Children's Cancer and Blood Disorders Centre, Boston, Massachusetts, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, and the Broad Institute, Dana-Farber/Boston Children's Cancer and Blood Disorders Centre, Boston, Massachusetts, USA
| | | | - Sabine Mueller
- Department of Neurology, Pediatrics and Neurosurgery, University of California San Francisco, San Francisco, California, USA
| | - Theo Nicolaides
- Department of Neurology, Pediatrics and Neurosurgery, University of California San Francisco, San Francisco, California, USA
| | - Giorgio Perilongo
- Department of Woman's and Child's Health, University of Padua, Padua, Italy
| | - Uri Tabori
- Division of Haematology/Oncology, Research Institute and The Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Gilbert Vezina
- Gilbert Family Neurofibromatosis Institute, Washington, District of Columbia, USA
- Brain Tumor Institute, Washington, District of Columbia, USA
- Division of Neuroradiology, Washington, District of Columbia, USA
| | - Katherine Warren
- National Cancer Institute, Pediatric Oncology and Neuro-Oncology Branches, Bethesda, Maryland, USA
| | - Olaf Witt
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yuan Zhu
- Center for Neuroscience and Behavioral Medicine, Washington, District of Columbia, USA
- Gilbert Family Neurofibromatosis Institute, Washington, District of Columbia, USA
- Center for Cancer and Immunology Research, Washington, District of Columbia, USA
| | - David T Jones
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mark Kieran
- Brain Tumor Center, Brain Tumor Translational Research, UC Department of Pediatrics, Cincinnati, Ohio, USA
| |
Collapse
|
154
|
Jittapiromsak N, Hou P, Liu HL, Sun J, Slopis JM, Chi TL. Prognostic Role of Conventional and Dynamic Contrast-Enhanced MRI in Optic Pathway Gliomas. J Neuroimaging 2017; 27:594-601. [DOI: 10.1111/jon.12450] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 04/26/2017] [Indexed: 12/12/2022] Open
Affiliation(s)
- Nutchawan Jittapiromsak
- Department of Diagnostic Radiology; The University of Texas MD Anderson Cancer Center; Houston TX
- Department of Radiology, Faculty of Medicine; Chulalongkorn University and the King Chulalongkorn Memorial Hospital; Pathumwan Bangkok Thailand
| | - Ping Hou
- Department of Imaging Physics; The University of Texas MD Anderson Cancer Center; Houston TX
| | - Ho-Ling Liu
- Department of Imaging Physics; The University of Texas MD Anderson Cancer Center; Houston TX
| | - Jia Sun
- Department of Biostatistics; The University of Texas MD Anderson Cancer Center; Houston TX
| | - John M. Slopis
- Department of Neuro-Oncology; The University of Texas MD Anderson Cancer Center; Houston TX
| | - T. Linda Chi
- Department of Diagnostic Radiology; The University of Texas MD Anderson Cancer Center; Houston TX
| |
Collapse
|
155
|
Abstract
Neoplasms in the central (CNS) and peripheral nervous system (PNS) in hereditary tumor syndromes play an important role in the neuropathological diagnostics. The benign and malignant PNS and CNS tumors that occur in the frequent neurofibromatosis type 1 (NF1) and type 2 (NF2) often represent essential factors for the course of the disease in those affected. Furthermore, certain clinical constellations (e.g. bilateral schwannomas of the auditory nerve, schwannomas at a young age and multiple meningiomas) can be important indications for a previously undiagnosed hereditary tumor disease. Other tumors occur practically regularly in association with certain germline defects, e.g. subependymal giant cell astrocytoma (SEGA) in tuberous sclerosis and dysplastic gangliocytoma of the cerebellum in Cowden's syndrome and can be indications in the diagnostics for an extended genetic counselling. This is not only important because many germline defects are based on new mutations, but also for the now established targeted therapy of certain tumors, e.g. inhibition of the mammalian target of rapamycin (mTOR) signaling pathway using temsirolimus for SEGA. Furthermore, knowledge about the possible constellations of genetic mosaics in hereditary tumor syndromes with the resulting (incomplete) syndrome manifestations is useful. This review article summarizes the most important hereditary tumor syndromes with involvement of the PNS and CNS.
Collapse
Affiliation(s)
- C Mawrin
- Institut für Neuropathologie, Otto-von-Guericke-Universität Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Deutschland.
| |
Collapse
|
156
|
Hanemann CO, Blakeley JO, Nunes FP, Robertson K, Stemmer-Rachamimov A, Mautner V, Kurtz A, Ferguson M, Widemann BC, Evans DG, Ferner R, Carroll SL, Korf B, Wolkenstein P, Knight P, Plotkin SR. Current status and recommendations for biomarkers and biobanking in neurofibromatosis. Neurology 2017; 87:S40-8. [PMID: 27527649 DOI: 10.1212/wnl.0000000000002932] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 03/30/2016] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE Clinically validated biomarkers for neurofibromatosis 1 (NF1), neurofibromatosis 2 (NF2), and schwannomatosis (SWN) have not been identified to date. The biomarker working group's goals are to (1) define biomarker needs in NF1, NF2, and SWN; (2) summarize existing data on biomarkers in NF1, NF2, and SWN; (3) outline recommendations for sample collection and biomarker development; and (4) standardize sample collection and methodology protocols where possible to promote comparison between studies by publishing standard operating procedures (SOPs). METHODS The biomarker group reviewed published data on biomarkers in NF1, NF2, and SWN and on biobanking efforts outside these diseases via literature search, defined the need for biomarkers in NF, and developed recommendations in a series of consensus meetings. RESULTS We describe existing biomarkers in NF and report consensus recommendations for SOP and a minimal clinical dataset to accompany samples derived from patients with NF1, NF2, and SWN in decentralized biobanks. CONCLUSIONS These recommendations are intended to provide clinicians and researchers with a common set of guidelines to collect and store biospecimens and for establishment of biobanks for NF1, NF2, and SWN.
Collapse
Affiliation(s)
- C Oliver Hanemann
- From Plymouth University (C.O.H.), Peninsula Schools of Medicine and Dentistry, The Institute of Translational and Stratified Medicine, Plymouth, UK; Department of Neurology (J.O.B.), Johns Hopkins University Medical School, Baltimore, MD; Department of Pediatrics (F.P.N.) and Department of Pediatrics, School of Medicine (K.R., M.F.), Indiana University; Tailored Therapeutics (F.P.N.), Eli Lilly and Company, Indianapolis, IN; Department of Pathology (A.S.-R.), Neuro-oncology (S.R.P.), Massachusetts General Hospital, Boston; Neurologische Klinik (V.M.), Uniklinik Eppendorf, Hamburg; Berlin-Brandenburg Center for Regenerative Therapies (A.K.), Charité Universitätsmedizin Berlin, Germany; Seoul National University (A.K.), College of Veterinary Medicine and Research Institute for Veterinary Science, Republic of Korea; NCI (B.C.W.), Pediatric Oncology Branch, Bethesda, MD; Genomic Medicine (D.G.E.), University of Manchester, UK; National Neurofibromatosis Service (R.F.), Department of Neurology, Guy's and St. Thomas' NHS Foundation Trust, London UK; Department of Pathology and Laboratory Medicine (S.L.C.), Medical University of South Carolina, Charleston; and Heflin Center for Genomic Sciences (B.K.), University of Alabama at Birmingham; Dermatology (P.W.), GHU Henri Mondor, Paris, France; Children's Tumor Foundation (P.K.), New York.
| | - Jaishri O Blakeley
- From Plymouth University (C.O.H.), Peninsula Schools of Medicine and Dentistry, The Institute of Translational and Stratified Medicine, Plymouth, UK; Department of Neurology (J.O.B.), Johns Hopkins University Medical School, Baltimore, MD; Department of Pediatrics (F.P.N.) and Department of Pediatrics, School of Medicine (K.R., M.F.), Indiana University; Tailored Therapeutics (F.P.N.), Eli Lilly and Company, Indianapolis, IN; Department of Pathology (A.S.-R.), Neuro-oncology (S.R.P.), Massachusetts General Hospital, Boston; Neurologische Klinik (V.M.), Uniklinik Eppendorf, Hamburg; Berlin-Brandenburg Center for Regenerative Therapies (A.K.), Charité Universitätsmedizin Berlin, Germany; Seoul National University (A.K.), College of Veterinary Medicine and Research Institute for Veterinary Science, Republic of Korea; NCI (B.C.W.), Pediatric Oncology Branch, Bethesda, MD; Genomic Medicine (D.G.E.), University of Manchester, UK; National Neurofibromatosis Service (R.F.), Department of Neurology, Guy's and St. Thomas' NHS Foundation Trust, London UK; Department of Pathology and Laboratory Medicine (S.L.C.), Medical University of South Carolina, Charleston; and Heflin Center for Genomic Sciences (B.K.), University of Alabama at Birmingham; Dermatology (P.W.), GHU Henri Mondor, Paris, France; Children's Tumor Foundation (P.K.), New York
| | - Fabio P Nunes
- From Plymouth University (C.O.H.), Peninsula Schools of Medicine and Dentistry, The Institute of Translational and Stratified Medicine, Plymouth, UK; Department of Neurology (J.O.B.), Johns Hopkins University Medical School, Baltimore, MD; Department of Pediatrics (F.P.N.) and Department of Pediatrics, School of Medicine (K.R., M.F.), Indiana University; Tailored Therapeutics (F.P.N.), Eli Lilly and Company, Indianapolis, IN; Department of Pathology (A.S.-R.), Neuro-oncology (S.R.P.), Massachusetts General Hospital, Boston; Neurologische Klinik (V.M.), Uniklinik Eppendorf, Hamburg; Berlin-Brandenburg Center for Regenerative Therapies (A.K.), Charité Universitätsmedizin Berlin, Germany; Seoul National University (A.K.), College of Veterinary Medicine and Research Institute for Veterinary Science, Republic of Korea; NCI (B.C.W.), Pediatric Oncology Branch, Bethesda, MD; Genomic Medicine (D.G.E.), University of Manchester, UK; National Neurofibromatosis Service (R.F.), Department of Neurology, Guy's and St. Thomas' NHS Foundation Trust, London UK; Department of Pathology and Laboratory Medicine (S.L.C.), Medical University of South Carolina, Charleston; and Heflin Center for Genomic Sciences (B.K.), University of Alabama at Birmingham; Dermatology (P.W.), GHU Henri Mondor, Paris, France; Children's Tumor Foundation (P.K.), New York
| | - Kent Robertson
- From Plymouth University (C.O.H.), Peninsula Schools of Medicine and Dentistry, The Institute of Translational and Stratified Medicine, Plymouth, UK; Department of Neurology (J.O.B.), Johns Hopkins University Medical School, Baltimore, MD; Department of Pediatrics (F.P.N.) and Department of Pediatrics, School of Medicine (K.R., M.F.), Indiana University; Tailored Therapeutics (F.P.N.), Eli Lilly and Company, Indianapolis, IN; Department of Pathology (A.S.-R.), Neuro-oncology (S.R.P.), Massachusetts General Hospital, Boston; Neurologische Klinik (V.M.), Uniklinik Eppendorf, Hamburg; Berlin-Brandenburg Center for Regenerative Therapies (A.K.), Charité Universitätsmedizin Berlin, Germany; Seoul National University (A.K.), College of Veterinary Medicine and Research Institute for Veterinary Science, Republic of Korea; NCI (B.C.W.), Pediatric Oncology Branch, Bethesda, MD; Genomic Medicine (D.G.E.), University of Manchester, UK; National Neurofibromatosis Service (R.F.), Department of Neurology, Guy's and St. Thomas' NHS Foundation Trust, London UK; Department of Pathology and Laboratory Medicine (S.L.C.), Medical University of South Carolina, Charleston; and Heflin Center for Genomic Sciences (B.K.), University of Alabama at Birmingham; Dermatology (P.W.), GHU Henri Mondor, Paris, France; Children's Tumor Foundation (P.K.), New York
| | - Anat Stemmer-Rachamimov
- From Plymouth University (C.O.H.), Peninsula Schools of Medicine and Dentistry, The Institute of Translational and Stratified Medicine, Plymouth, UK; Department of Neurology (J.O.B.), Johns Hopkins University Medical School, Baltimore, MD; Department of Pediatrics (F.P.N.) and Department of Pediatrics, School of Medicine (K.R., M.F.), Indiana University; Tailored Therapeutics (F.P.N.), Eli Lilly and Company, Indianapolis, IN; Department of Pathology (A.S.-R.), Neuro-oncology (S.R.P.), Massachusetts General Hospital, Boston; Neurologische Klinik (V.M.), Uniklinik Eppendorf, Hamburg; Berlin-Brandenburg Center for Regenerative Therapies (A.K.), Charité Universitätsmedizin Berlin, Germany; Seoul National University (A.K.), College of Veterinary Medicine and Research Institute for Veterinary Science, Republic of Korea; NCI (B.C.W.), Pediatric Oncology Branch, Bethesda, MD; Genomic Medicine (D.G.E.), University of Manchester, UK; National Neurofibromatosis Service (R.F.), Department of Neurology, Guy's and St. Thomas' NHS Foundation Trust, London UK; Department of Pathology and Laboratory Medicine (S.L.C.), Medical University of South Carolina, Charleston; and Heflin Center for Genomic Sciences (B.K.), University of Alabama at Birmingham; Dermatology (P.W.), GHU Henri Mondor, Paris, France; Children's Tumor Foundation (P.K.), New York
| | - Victor Mautner
- From Plymouth University (C.O.H.), Peninsula Schools of Medicine and Dentistry, The Institute of Translational and Stratified Medicine, Plymouth, UK; Department of Neurology (J.O.B.), Johns Hopkins University Medical School, Baltimore, MD; Department of Pediatrics (F.P.N.) and Department of Pediatrics, School of Medicine (K.R., M.F.), Indiana University; Tailored Therapeutics (F.P.N.), Eli Lilly and Company, Indianapolis, IN; Department of Pathology (A.S.-R.), Neuro-oncology (S.R.P.), Massachusetts General Hospital, Boston; Neurologische Klinik (V.M.), Uniklinik Eppendorf, Hamburg; Berlin-Brandenburg Center for Regenerative Therapies (A.K.), Charité Universitätsmedizin Berlin, Germany; Seoul National University (A.K.), College of Veterinary Medicine and Research Institute for Veterinary Science, Republic of Korea; NCI (B.C.W.), Pediatric Oncology Branch, Bethesda, MD; Genomic Medicine (D.G.E.), University of Manchester, UK; National Neurofibromatosis Service (R.F.), Department of Neurology, Guy's and St. Thomas' NHS Foundation Trust, London UK; Department of Pathology and Laboratory Medicine (S.L.C.), Medical University of South Carolina, Charleston; and Heflin Center for Genomic Sciences (B.K.), University of Alabama at Birmingham; Dermatology (P.W.), GHU Henri Mondor, Paris, France; Children's Tumor Foundation (P.K.), New York
| | - Andreas Kurtz
- From Plymouth University (C.O.H.), Peninsula Schools of Medicine and Dentistry, The Institute of Translational and Stratified Medicine, Plymouth, UK; Department of Neurology (J.O.B.), Johns Hopkins University Medical School, Baltimore, MD; Department of Pediatrics (F.P.N.) and Department of Pediatrics, School of Medicine (K.R., M.F.), Indiana University; Tailored Therapeutics (F.P.N.), Eli Lilly and Company, Indianapolis, IN; Department of Pathology (A.S.-R.), Neuro-oncology (S.R.P.), Massachusetts General Hospital, Boston; Neurologische Klinik (V.M.), Uniklinik Eppendorf, Hamburg; Berlin-Brandenburg Center for Regenerative Therapies (A.K.), Charité Universitätsmedizin Berlin, Germany; Seoul National University (A.K.), College of Veterinary Medicine and Research Institute for Veterinary Science, Republic of Korea; NCI (B.C.W.), Pediatric Oncology Branch, Bethesda, MD; Genomic Medicine (D.G.E.), University of Manchester, UK; National Neurofibromatosis Service (R.F.), Department of Neurology, Guy's and St. Thomas' NHS Foundation Trust, London UK; Department of Pathology and Laboratory Medicine (S.L.C.), Medical University of South Carolina, Charleston; and Heflin Center for Genomic Sciences (B.K.), University of Alabama at Birmingham; Dermatology (P.W.), GHU Henri Mondor, Paris, France; Children's Tumor Foundation (P.K.), New York
| | - Michael Ferguson
- From Plymouth University (C.O.H.), Peninsula Schools of Medicine and Dentistry, The Institute of Translational and Stratified Medicine, Plymouth, UK; Department of Neurology (J.O.B.), Johns Hopkins University Medical School, Baltimore, MD; Department of Pediatrics (F.P.N.) and Department of Pediatrics, School of Medicine (K.R., M.F.), Indiana University; Tailored Therapeutics (F.P.N.), Eli Lilly and Company, Indianapolis, IN; Department of Pathology (A.S.-R.), Neuro-oncology (S.R.P.), Massachusetts General Hospital, Boston; Neurologische Klinik (V.M.), Uniklinik Eppendorf, Hamburg; Berlin-Brandenburg Center for Regenerative Therapies (A.K.), Charité Universitätsmedizin Berlin, Germany; Seoul National University (A.K.), College of Veterinary Medicine and Research Institute for Veterinary Science, Republic of Korea; NCI (B.C.W.), Pediatric Oncology Branch, Bethesda, MD; Genomic Medicine (D.G.E.), University of Manchester, UK; National Neurofibromatosis Service (R.F.), Department of Neurology, Guy's and St. Thomas' NHS Foundation Trust, London UK; Department of Pathology and Laboratory Medicine (S.L.C.), Medical University of South Carolina, Charleston; and Heflin Center for Genomic Sciences (B.K.), University of Alabama at Birmingham; Dermatology (P.W.), GHU Henri Mondor, Paris, France; Children's Tumor Foundation (P.K.), New York
| | - Brigitte C Widemann
- From Plymouth University (C.O.H.), Peninsula Schools of Medicine and Dentistry, The Institute of Translational and Stratified Medicine, Plymouth, UK; Department of Neurology (J.O.B.), Johns Hopkins University Medical School, Baltimore, MD; Department of Pediatrics (F.P.N.) and Department of Pediatrics, School of Medicine (K.R., M.F.), Indiana University; Tailored Therapeutics (F.P.N.), Eli Lilly and Company, Indianapolis, IN; Department of Pathology (A.S.-R.), Neuro-oncology (S.R.P.), Massachusetts General Hospital, Boston; Neurologische Klinik (V.M.), Uniklinik Eppendorf, Hamburg; Berlin-Brandenburg Center for Regenerative Therapies (A.K.), Charité Universitätsmedizin Berlin, Germany; Seoul National University (A.K.), College of Veterinary Medicine and Research Institute for Veterinary Science, Republic of Korea; NCI (B.C.W.), Pediatric Oncology Branch, Bethesda, MD; Genomic Medicine (D.G.E.), University of Manchester, UK; National Neurofibromatosis Service (R.F.), Department of Neurology, Guy's and St. Thomas' NHS Foundation Trust, London UK; Department of Pathology and Laboratory Medicine (S.L.C.), Medical University of South Carolina, Charleston; and Heflin Center for Genomic Sciences (B.K.), University of Alabama at Birmingham; Dermatology (P.W.), GHU Henri Mondor, Paris, France; Children's Tumor Foundation (P.K.), New York
| | - D Gareth Evans
- From Plymouth University (C.O.H.), Peninsula Schools of Medicine and Dentistry, The Institute of Translational and Stratified Medicine, Plymouth, UK; Department of Neurology (J.O.B.), Johns Hopkins University Medical School, Baltimore, MD; Department of Pediatrics (F.P.N.) and Department of Pediatrics, School of Medicine (K.R., M.F.), Indiana University; Tailored Therapeutics (F.P.N.), Eli Lilly and Company, Indianapolis, IN; Department of Pathology (A.S.-R.), Neuro-oncology (S.R.P.), Massachusetts General Hospital, Boston; Neurologische Klinik (V.M.), Uniklinik Eppendorf, Hamburg; Berlin-Brandenburg Center for Regenerative Therapies (A.K.), Charité Universitätsmedizin Berlin, Germany; Seoul National University (A.K.), College of Veterinary Medicine and Research Institute for Veterinary Science, Republic of Korea; NCI (B.C.W.), Pediatric Oncology Branch, Bethesda, MD; Genomic Medicine (D.G.E.), University of Manchester, UK; National Neurofibromatosis Service (R.F.), Department of Neurology, Guy's and St. Thomas' NHS Foundation Trust, London UK; Department of Pathology and Laboratory Medicine (S.L.C.), Medical University of South Carolina, Charleston; and Heflin Center for Genomic Sciences (B.K.), University of Alabama at Birmingham; Dermatology (P.W.), GHU Henri Mondor, Paris, France; Children's Tumor Foundation (P.K.), New York
| | - Rosalie Ferner
- From Plymouth University (C.O.H.), Peninsula Schools of Medicine and Dentistry, The Institute of Translational and Stratified Medicine, Plymouth, UK; Department of Neurology (J.O.B.), Johns Hopkins University Medical School, Baltimore, MD; Department of Pediatrics (F.P.N.) and Department of Pediatrics, School of Medicine (K.R., M.F.), Indiana University; Tailored Therapeutics (F.P.N.), Eli Lilly and Company, Indianapolis, IN; Department of Pathology (A.S.-R.), Neuro-oncology (S.R.P.), Massachusetts General Hospital, Boston; Neurologische Klinik (V.M.), Uniklinik Eppendorf, Hamburg; Berlin-Brandenburg Center for Regenerative Therapies (A.K.), Charité Universitätsmedizin Berlin, Germany; Seoul National University (A.K.), College of Veterinary Medicine and Research Institute for Veterinary Science, Republic of Korea; NCI (B.C.W.), Pediatric Oncology Branch, Bethesda, MD; Genomic Medicine (D.G.E.), University of Manchester, UK; National Neurofibromatosis Service (R.F.), Department of Neurology, Guy's and St. Thomas' NHS Foundation Trust, London UK; Department of Pathology and Laboratory Medicine (S.L.C.), Medical University of South Carolina, Charleston; and Heflin Center for Genomic Sciences (B.K.), University of Alabama at Birmingham; Dermatology (P.W.), GHU Henri Mondor, Paris, France; Children's Tumor Foundation (P.K.), New York
| | - Steven L Carroll
- From Plymouth University (C.O.H.), Peninsula Schools of Medicine and Dentistry, The Institute of Translational and Stratified Medicine, Plymouth, UK; Department of Neurology (J.O.B.), Johns Hopkins University Medical School, Baltimore, MD; Department of Pediatrics (F.P.N.) and Department of Pediatrics, School of Medicine (K.R., M.F.), Indiana University; Tailored Therapeutics (F.P.N.), Eli Lilly and Company, Indianapolis, IN; Department of Pathology (A.S.-R.), Neuro-oncology (S.R.P.), Massachusetts General Hospital, Boston; Neurologische Klinik (V.M.), Uniklinik Eppendorf, Hamburg; Berlin-Brandenburg Center for Regenerative Therapies (A.K.), Charité Universitätsmedizin Berlin, Germany; Seoul National University (A.K.), College of Veterinary Medicine and Research Institute for Veterinary Science, Republic of Korea; NCI (B.C.W.), Pediatric Oncology Branch, Bethesda, MD; Genomic Medicine (D.G.E.), University of Manchester, UK; National Neurofibromatosis Service (R.F.), Department of Neurology, Guy's and St. Thomas' NHS Foundation Trust, London UK; Department of Pathology and Laboratory Medicine (S.L.C.), Medical University of South Carolina, Charleston; and Heflin Center for Genomic Sciences (B.K.), University of Alabama at Birmingham; Dermatology (P.W.), GHU Henri Mondor, Paris, France; Children's Tumor Foundation (P.K.), New York
| | - Bruce Korf
- From Plymouth University (C.O.H.), Peninsula Schools of Medicine and Dentistry, The Institute of Translational and Stratified Medicine, Plymouth, UK; Department of Neurology (J.O.B.), Johns Hopkins University Medical School, Baltimore, MD; Department of Pediatrics (F.P.N.) and Department of Pediatrics, School of Medicine (K.R., M.F.), Indiana University; Tailored Therapeutics (F.P.N.), Eli Lilly and Company, Indianapolis, IN; Department of Pathology (A.S.-R.), Neuro-oncology (S.R.P.), Massachusetts General Hospital, Boston; Neurologische Klinik (V.M.), Uniklinik Eppendorf, Hamburg; Berlin-Brandenburg Center for Regenerative Therapies (A.K.), Charité Universitätsmedizin Berlin, Germany; Seoul National University (A.K.), College of Veterinary Medicine and Research Institute for Veterinary Science, Republic of Korea; NCI (B.C.W.), Pediatric Oncology Branch, Bethesda, MD; Genomic Medicine (D.G.E.), University of Manchester, UK; National Neurofibromatosis Service (R.F.), Department of Neurology, Guy's and St. Thomas' NHS Foundation Trust, London UK; Department of Pathology and Laboratory Medicine (S.L.C.), Medical University of South Carolina, Charleston; and Heflin Center for Genomic Sciences (B.K.), University of Alabama at Birmingham; Dermatology (P.W.), GHU Henri Mondor, Paris, France; Children's Tumor Foundation (P.K.), New York
| | - Pierre Wolkenstein
- From Plymouth University (C.O.H.), Peninsula Schools of Medicine and Dentistry, The Institute of Translational and Stratified Medicine, Plymouth, UK; Department of Neurology (J.O.B.), Johns Hopkins University Medical School, Baltimore, MD; Department of Pediatrics (F.P.N.) and Department of Pediatrics, School of Medicine (K.R., M.F.), Indiana University; Tailored Therapeutics (F.P.N.), Eli Lilly and Company, Indianapolis, IN; Department of Pathology (A.S.-R.), Neuro-oncology (S.R.P.), Massachusetts General Hospital, Boston; Neurologische Klinik (V.M.), Uniklinik Eppendorf, Hamburg; Berlin-Brandenburg Center for Regenerative Therapies (A.K.), Charité Universitätsmedizin Berlin, Germany; Seoul National University (A.K.), College of Veterinary Medicine and Research Institute for Veterinary Science, Republic of Korea; NCI (B.C.W.), Pediatric Oncology Branch, Bethesda, MD; Genomic Medicine (D.G.E.), University of Manchester, UK; National Neurofibromatosis Service (R.F.), Department of Neurology, Guy's and St. Thomas' NHS Foundation Trust, London UK; Department of Pathology and Laboratory Medicine (S.L.C.), Medical University of South Carolina, Charleston; and Heflin Center for Genomic Sciences (B.K.), University of Alabama at Birmingham; Dermatology (P.W.), GHU Henri Mondor, Paris, France; Children's Tumor Foundation (P.K.), New York
| | - Pamela Knight
- From Plymouth University (C.O.H.), Peninsula Schools of Medicine and Dentistry, The Institute of Translational and Stratified Medicine, Plymouth, UK; Department of Neurology (J.O.B.), Johns Hopkins University Medical School, Baltimore, MD; Department of Pediatrics (F.P.N.) and Department of Pediatrics, School of Medicine (K.R., M.F.), Indiana University; Tailored Therapeutics (F.P.N.), Eli Lilly and Company, Indianapolis, IN; Department of Pathology (A.S.-R.), Neuro-oncology (S.R.P.), Massachusetts General Hospital, Boston; Neurologische Klinik (V.M.), Uniklinik Eppendorf, Hamburg; Berlin-Brandenburg Center for Regenerative Therapies (A.K.), Charité Universitätsmedizin Berlin, Germany; Seoul National University (A.K.), College of Veterinary Medicine and Research Institute for Veterinary Science, Republic of Korea; NCI (B.C.W.), Pediatric Oncology Branch, Bethesda, MD; Genomic Medicine (D.G.E.), University of Manchester, UK; National Neurofibromatosis Service (R.F.), Department of Neurology, Guy's and St. Thomas' NHS Foundation Trust, London UK; Department of Pathology and Laboratory Medicine (S.L.C.), Medical University of South Carolina, Charleston; and Heflin Center for Genomic Sciences (B.K.), University of Alabama at Birmingham; Dermatology (P.W.), GHU Henri Mondor, Paris, France; Children's Tumor Foundation (P.K.), New York
| | - Scott R Plotkin
- From Plymouth University (C.O.H.), Peninsula Schools of Medicine and Dentistry, The Institute of Translational and Stratified Medicine, Plymouth, UK; Department of Neurology (J.O.B.), Johns Hopkins University Medical School, Baltimore, MD; Department of Pediatrics (F.P.N.) and Department of Pediatrics, School of Medicine (K.R., M.F.), Indiana University; Tailored Therapeutics (F.P.N.), Eli Lilly and Company, Indianapolis, IN; Department of Pathology (A.S.-R.), Neuro-oncology (S.R.P.), Massachusetts General Hospital, Boston; Neurologische Klinik (V.M.), Uniklinik Eppendorf, Hamburg; Berlin-Brandenburg Center for Regenerative Therapies (A.K.), Charité Universitätsmedizin Berlin, Germany; Seoul National University (A.K.), College of Veterinary Medicine and Research Institute for Veterinary Science, Republic of Korea; NCI (B.C.W.), Pediatric Oncology Branch, Bethesda, MD; Genomic Medicine (D.G.E.), University of Manchester, UK; National Neurofibromatosis Service (R.F.), Department of Neurology, Guy's and St. Thomas' NHS Foundation Trust, London UK; Department of Pathology and Laboratory Medicine (S.L.C.), Medical University of South Carolina, Charleston; and Heflin Center for Genomic Sciences (B.K.), University of Alabama at Birmingham; Dermatology (P.W.), GHU Henri Mondor, Paris, France; Children's Tumor Foundation (P.K.), New York
| | | |
Collapse
|
157
|
|
158
|
Abstract
Children with neurofibromatosis type 1 frequently manifest optic pathway gliomas-low-grade gliomas intrinsic to the visual pathway. This review describes the molecular and genetic mechanisms driving optic pathway gliomas as well as the clinical symptoms of this relatively common genetic condition. Recommendations for clinical management and descriptions of the newest imaging techniques are discussed.
Collapse
Affiliation(s)
| | - Robert A Avery
- Division of Ophthalmology, The Children's Hospital of Philadelphia, Philadelphia, PA; Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
159
|
Mahdi J, Shah AC, Sato A, Morris SM, McKinstry RC, Listernick R, Packer RJ, Fisher MJ, Gutmann DH. A multi-institutional study of brainstem gliomas in children with neurofibromatosis type 1. Neurology 2017; 88:1584-1589. [PMID: 28330960 DOI: 10.1212/wnl.0000000000003881] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 01/24/2017] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To define the clinical and radiologic features of brainstem gliomas (BSGs) in children with neurofibromatosis type 1 (NF1). METHODS We performed a retrospective cross-sectional study of 133 children with NF1 and concurrent BSGs cared for at 4 NF1 referral centers. BSG was determined using radiographic criteria. Age at diagnosis, tumor location and appearance, clinical symptoms, treatment, and presence of a concurrent optic pathway glioma were assessed. RESULTS The average age at BSG diagnosis was 7.2 years, and tumors occurred most often in the midbrain and medulla (66%). The majority of children with NF1-BSGs were asymptomatic (54%) and were not treated (88%). Only 9 of the 72 asymptomatic children received treatment because of progressive tumor enlargement. In contrast, 61 children presented with clinical signs/symptoms attributable to their BSG; these individuals were older and more often had focal lesions. Thirty-one patients underwent treatment for their tumor, and 14 received CSF diversion only. Progression-free survival was ∼3 years shorter for children receiving tumor-directed therapy relative to those who had either no treatment or CSF diversion only. Overall survival was 85% for the tumor-directed therapy group, whereas no deaths were reported in the untreated or CSF diversion groups. CONCLUSIONS Unlike children with sporadically occurring BSGs, most children with NF1-BSGs were asymptomatic, and few individuals died from complications of their tumor. Those requiring tumor-directed treatment tended to be older children with focal lesions, and had clinically more aggressive disease relative to those who were not treated or underwent CSF diversion only.
Collapse
Affiliation(s)
- Jasia Mahdi
- From the Departments of Neurology (J.M., S.M.M., D.H.G.) and Radiology (R.C.M.), Washington University School of Medicine, St. Louis, MO; Division of Oncology (A.C.S., M.J.F.), Children's Hospital of Philadelphia, PA; Center for Neuroscience of Behavioral Medicine (A.S., R.J.P.), Children's National Medical Center, Washington, DC; Division of Academic General Pediatrics (R.L.), Feinberg School of Medicine, Northwestern University, Ann & Robert H. Lurie Children's Hospital of Chicago, IL; and Department of Pediatrics (M.J.F.), The Perelman School of Medicine at The University of Pennsylvania, Philadelphia
| | - Amish C Shah
- From the Departments of Neurology (J.M., S.M.M., D.H.G.) and Radiology (R.C.M.), Washington University School of Medicine, St. Louis, MO; Division of Oncology (A.C.S., M.J.F.), Children's Hospital of Philadelphia, PA; Center for Neuroscience of Behavioral Medicine (A.S., R.J.P.), Children's National Medical Center, Washington, DC; Division of Academic General Pediatrics (R.L.), Feinberg School of Medicine, Northwestern University, Ann & Robert H. Lurie Children's Hospital of Chicago, IL; and Department of Pediatrics (M.J.F.), The Perelman School of Medicine at The University of Pennsylvania, Philadelphia
| | - Aimee Sato
- From the Departments of Neurology (J.M., S.M.M., D.H.G.) and Radiology (R.C.M.), Washington University School of Medicine, St. Louis, MO; Division of Oncology (A.C.S., M.J.F.), Children's Hospital of Philadelphia, PA; Center for Neuroscience of Behavioral Medicine (A.S., R.J.P.), Children's National Medical Center, Washington, DC; Division of Academic General Pediatrics (R.L.), Feinberg School of Medicine, Northwestern University, Ann & Robert H. Lurie Children's Hospital of Chicago, IL; and Department of Pediatrics (M.J.F.), The Perelman School of Medicine at The University of Pennsylvania, Philadelphia
| | - Stephanie M Morris
- From the Departments of Neurology (J.M., S.M.M., D.H.G.) and Radiology (R.C.M.), Washington University School of Medicine, St. Louis, MO; Division of Oncology (A.C.S., M.J.F.), Children's Hospital of Philadelphia, PA; Center for Neuroscience of Behavioral Medicine (A.S., R.J.P.), Children's National Medical Center, Washington, DC; Division of Academic General Pediatrics (R.L.), Feinberg School of Medicine, Northwestern University, Ann & Robert H. Lurie Children's Hospital of Chicago, IL; and Department of Pediatrics (M.J.F.), The Perelman School of Medicine at The University of Pennsylvania, Philadelphia
| | - Robert C McKinstry
- From the Departments of Neurology (J.M., S.M.M., D.H.G.) and Radiology (R.C.M.), Washington University School of Medicine, St. Louis, MO; Division of Oncology (A.C.S., M.J.F.), Children's Hospital of Philadelphia, PA; Center for Neuroscience of Behavioral Medicine (A.S., R.J.P.), Children's National Medical Center, Washington, DC; Division of Academic General Pediatrics (R.L.), Feinberg School of Medicine, Northwestern University, Ann & Robert H. Lurie Children's Hospital of Chicago, IL; and Department of Pediatrics (M.J.F.), The Perelman School of Medicine at The University of Pennsylvania, Philadelphia
| | - Robert Listernick
- From the Departments of Neurology (J.M., S.M.M., D.H.G.) and Radiology (R.C.M.), Washington University School of Medicine, St. Louis, MO; Division of Oncology (A.C.S., M.J.F.), Children's Hospital of Philadelphia, PA; Center for Neuroscience of Behavioral Medicine (A.S., R.J.P.), Children's National Medical Center, Washington, DC; Division of Academic General Pediatrics (R.L.), Feinberg School of Medicine, Northwestern University, Ann & Robert H. Lurie Children's Hospital of Chicago, IL; and Department of Pediatrics (M.J.F.), The Perelman School of Medicine at The University of Pennsylvania, Philadelphia
| | - Roger J Packer
- From the Departments of Neurology (J.M., S.M.M., D.H.G.) and Radiology (R.C.M.), Washington University School of Medicine, St. Louis, MO; Division of Oncology (A.C.S., M.J.F.), Children's Hospital of Philadelphia, PA; Center for Neuroscience of Behavioral Medicine (A.S., R.J.P.), Children's National Medical Center, Washington, DC; Division of Academic General Pediatrics (R.L.), Feinberg School of Medicine, Northwestern University, Ann & Robert H. Lurie Children's Hospital of Chicago, IL; and Department of Pediatrics (M.J.F.), The Perelman School of Medicine at The University of Pennsylvania, Philadelphia
| | - Michael J Fisher
- From the Departments of Neurology (J.M., S.M.M., D.H.G.) and Radiology (R.C.M.), Washington University School of Medicine, St. Louis, MO; Division of Oncology (A.C.S., M.J.F.), Children's Hospital of Philadelphia, PA; Center for Neuroscience of Behavioral Medicine (A.S., R.J.P.), Children's National Medical Center, Washington, DC; Division of Academic General Pediatrics (R.L.), Feinberg School of Medicine, Northwestern University, Ann & Robert H. Lurie Children's Hospital of Chicago, IL; and Department of Pediatrics (M.J.F.), The Perelman School of Medicine at The University of Pennsylvania, Philadelphia
| | - David H Gutmann
- From the Departments of Neurology (J.M., S.M.M., D.H.G.) and Radiology (R.C.M.), Washington University School of Medicine, St. Louis, MO; Division of Oncology (A.C.S., M.J.F.), Children's Hospital of Philadelphia, PA; Center for Neuroscience of Behavioral Medicine (A.S., R.J.P.), Children's National Medical Center, Washington, DC; Division of Academic General Pediatrics (R.L.), Feinberg School of Medicine, Northwestern University, Ann & Robert H. Lurie Children's Hospital of Chicago, IL; and Department of Pediatrics (M.J.F.), The Perelman School of Medicine at The University of Pennsylvania, Philadelphia.
| |
Collapse
|
160
|
Gutmann DH, Ferner RE, Listernick RH, Korf BR, Wolters PL, Johnson KJ. Neurofibromatosis type 1. Nat Rev Dis Primers 2017; 3:17004. [PMID: 28230061 DOI: 10.1038/nrdp.2017.4] [Citation(s) in RCA: 435] [Impact Index Per Article: 62.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Neurofibromatosis type 1 is a complex autosomal dominant disorder caused by germline mutations in the NF1 tumour suppressor gene. Nearly all individuals with neurofibromatosis type 1 develop pigmentary lesions (café-au-lait macules, skinfold freckling and Lisch nodules) and dermal neurofibromas. Some individuals develop skeletal abnormalities (scoliosis, tibial pseudarthrosis and orbital dysplasia), brain tumours (optic pathway gliomas and glioblastoma), peripheral nerve tumours (spinal neurofibromas, plexiform neurofibromas and malignant peripheral nerve sheath tumours), learning disabilities, attention deficits, and social and behavioural problems, which can negatively affect quality of life. With the identification of NF1 and the generation of accurate preclinical mouse strains that model some of these clinical features, therapies that target the underlying molecular and cellular pathophysiology for neurofibromatosis type 1 are becoming available. Although no single treatment exists, current clinical management strategies include early detection of disease phenotypes (risk assessment) and biologically targeted therapies. Similarly, new medical and behavioural interventions are emerging to improve the quality of life of patients. Although considerable progress has been made in understanding this condition, numerous challenges remain; a collaborative and interdisciplinary approach is required to manage individuals with neurofibromatosis type1 and to develop effective treatments.
Collapse
Affiliation(s)
- David H Gutmann
- Department of Neurology, Washington University School of Medicine, Box 8111, 660 S. Euclid Avenue, St. Louis, Missouri 63110, USA
| | - Rosalie E Ferner
- Department of Neurology, Guy's and St. Thomas' NHS Foundation Trust, London, UK.,Department of Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Robert H Listernick
- Department of Academic General Pediatrics and Primary Care, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA.,Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Bruce R Korf
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Pamela L Wolters
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | | |
Collapse
|
161
|
Vijapura C, Saad Aldin E, Capizzano AA, Policeni B, Sato Y, Moritani T. Genetic Syndromes Associated with Central Nervous System Tumors. Radiographics 2017; 37:258-280. [DOI: 10.1148/rg.2017160057] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
162
|
Gaudino S, Martucci M, Russo R, Visconti E, Gangemi E, D'Argento F, Verdolotti T, Lauriola L, Colosimo C. MR imaging of brain pilocytic astrocytoma: beyond the stereotype of benign astrocytoma. Childs Nerv Syst 2017; 33:35-54. [PMID: 27757570 DOI: 10.1007/s00381-016-3262-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 10/03/2016] [Indexed: 01/21/2023]
Abstract
BACKGROUND Pilocytic astrocytoma (PA) is the most common pediatric brain glioma and is considered the prototype of benign circumscribed astrocytoma. Despite its low malignancy, the CT and MRI features of brain PA may resemble those of much more aggressive brain tumors. Misdiagnosis of PA is particularly easy when it demonstrates MR morphological and non-morphological findings that are inconsistent with its non-aggressive nature and that overlap with the features of more aggressive brain tumors. METHOD Basing on the evidence that the variation in the histological, genetic, and metabolic "fingerprint" for brain PA is dependent on tumor location, and the hypothesis that tumor location is related to the broad spectrum of morphological and non-morphological MR imaging findings, the authors discuss the MR imaging appearance of brain PA using a location-based approach to underline the typical and less typical imaging features and the main differential diagnosis of brain PA. A brief summary of the main pathological and clinical features, the natural history, and the treatment of brain PA is also provided. RESULT A combination of morphological and non-morphological MR imaging features and a site-based approach to differential diagnosis are required for a pre-operative diagnosis. The new "cutting-edge" MR imaging sequences have the potential to impact the ease and confidence of pediatric brain tumor interpretation and offer a more efficient diagnostic work-up. CONCLUSIONS Although the typical imaging features of brain pilocytic astrocytoma make radiological diagnosis relatively easy, an atypical and more aggressive appearance can lead to misdiagnosis. Knowing the broad spectrum of imaging characteristics on conventional and advanced MR imaging is important for accurate pre-operative radiological diagnosis and correctly interpreting changes during follow-up.
Collapse
Affiliation(s)
- Simona Gaudino
- Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli, Largo A. Gemelli, 1, 00168, Rome, Italy.
| | - Matia Martucci
- Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli, Largo A. Gemelli, 1, 00168, Rome, Italy
| | - Rosellina Russo
- Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli, Largo A. Gemelli, 1, 00168, Rome, Italy
| | - Emiliano Visconti
- Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli, Largo A. Gemelli, 1, 00168, Rome, Italy
| | - Emma Gangemi
- Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli, Largo A. Gemelli, 1, 00168, Rome, Italy
| | - Francesco D'Argento
- Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli, Largo A. Gemelli, 1, 00168, Rome, Italy
| | - Tommaso Verdolotti
- Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli, Largo A. Gemelli, 1, 00168, Rome, Italy
| | - Libero Lauriola
- Institute of Pathological Anatomy, Fondazione Policlinico Universitario Agostino Gemelli, Largo A. Gemelli, 1, 00168, Rome, Italy
| | - Cesare Colosimo
- Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli, Largo A. Gemelli, 1, 00168, Rome, Italy
| |
Collapse
|
163
|
Karmakar S, Reilly KM. The role of the immune system in neurofibromatosis type 1-associated nervous system tumors. CNS Oncol 2016; 6:45-60. [PMID: 28001089 DOI: 10.2217/cns-2016-0024] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
With the recent development of new anticancer therapies targeting the immune system, it is important to understand which immune cell types and cytokines play critical roles in suppressing or promoting tumorigenesis. The role of mast cells in promoting neurofibroma growth in neurofibromatosis type 1 (NF1) patients was hypothesized decades ago. More recent experiments in mouse models have demonstrated the causal role of mast cells in neurofibroma development and of microglia in optic pathway glioma development. We review here what is known about the role of NF1 mutation in immune cell function and the role of immune cells in promoting tumorigenesis in NF1. We also review the therapies targeting immune cell pathways and their promise in NF1 tumors.
Collapse
Affiliation(s)
- Souvik Karmakar
- Rare Tumors Initiative, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Dr, Bethesda, MD 20814, USA
| | - Karlyne M Reilly
- Rare Tumors Initiative, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Dr, Bethesda, MD 20814, USA
| |
Collapse
|
164
|
Wagner MW, Poretti A, Benson JE, Huisman TAGM. Neuroimaging Findings in Pediatric Genetic Skeletal Disorders: A Review. J Neuroimaging 2016; 27:162-209. [PMID: 28000960 DOI: 10.1111/jon.12413] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 11/01/2016] [Indexed: 12/15/2022] Open
Abstract
Genetic skeletal disorders (GSDs) are a heterogeneous group characterized by an intrinsic abnormality in growth and (re-)modeling of cartilage and bone. A large subgroup of GSDs has additional involvement of other structures/organs beside the skeleton, such as the central nervous system (CNS). CNS abnormalities have an important role in long-term prognosis of children with GSDs and should consequently not be missed. Sensitive and specific identification of CNS lesions while evaluating a child with a GSD requires a detailed knowledge of the possible associated CNS abnormalities. Here, we provide a pattern-recognition approach for neuroimaging findings in GSDs guided by the obvious skeletal manifestations of GSD. In particular, we summarize which CNS findings should be ruled out with each GSD. The diseases (n = 180) are classified based on the skeletal involvement (1. abnormal metaphysis or epiphysis, 2. abnormal size/number of bones, 3. abnormal shape of bones and joints, and 4. abnormal dynamic or structural changes). For each disease, skeletal involvement was defined in accordance with Online Mendelian Inheritance in Man. Morphological CNS involvement has been described based on extensive literature search. Selected examples will be shown based on prevalence of the diseases and significance of the CNS involvement. CNS involvement is common in GSDs. A wide spectrum of morphological abnormalities is associated with GSDs. Early diagnosis of CNS involvement is important in the management of children with GSDs. This pattern-recognition approach aims to assist and guide physicians in the diagnostic work-up of CNS involvement in children with GSDs and their management.
Collapse
Affiliation(s)
- Matthias W Wagner
- Section of Pediatric Neuroradiology, Division of Pediatric Radiology, Russell H. Morgan Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, MD.,Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
| | - Andrea Poretti
- Section of Pediatric Neuroradiology, Division of Pediatric Radiology, Russell H. Morgan Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jane E Benson
- Section of Pediatric Neuroradiology, Division of Pediatric Radiology, Russell H. Morgan Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Thierry A G M Huisman
- Section of Pediatric Neuroradiology, Division of Pediatric Radiology, Russell H. Morgan Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
165
|
Toonen JA, Solga AC, Ma Y, Gutmann DH. Estrogen activation of microglia underlies the sexually dimorphic differences in Nf1 optic glioma-induced retinal pathology. J Exp Med 2016; 214:17-25. [PMID: 27923908 PMCID: PMC5206494 DOI: 10.1084/jem.20160447] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 09/23/2016] [Accepted: 11/11/2016] [Indexed: 01/15/2023] Open
Abstract
Children with neurofibromatosis type 1 (NF1) develop low-grade brain tumors throughout the optic pathway. Nearly 50% of children with optic pathway gliomas (OPGs) experience visual impairment, and few regain their vision after chemotherapy. Recent studies have revealed that girls with optic nerve gliomas are five times more likely to lose vision and require treatment than boys. To determine the mechanism underlying this sexually dimorphic difference in clinical outcome, we leveraged Nf1 optic glioma (Nf1-OPG) mice. We demonstrate that female Nf1-OPG mice exhibit greater retinal ganglion cell (RGC) loss and only females have retinal nerve fiber layer (RNFL) thinning, despite mice of both sexes harboring tumors of identical volumes and proliferation. Female gonadal sex hormones are responsible for this sexual dimorphism, as ovariectomy, but not castration, of Nf1-OPG mice normalizes RGC survival and RNFL thickness. In addition, female Nf1-OPG mice have threefold more microglia than their male counterparts, and minocycline inhibition of microglia corrects the retinal pathology. Moreover, pharmacologic inhibition of microglial estrogen receptor-β (ERβ) function corrects the retinal abnormalities in female Nf1-OPG mice. Collectively, these studies establish that female gonadal sex hormones underlie the sexual dimorphic differences in Nf1 optic glioma-induced retinal dysfunction by operating at the level of tumor-associated microglial activation.
Collapse
Affiliation(s)
- Joseph A Toonen
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| | - Anne C Solga
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| | - Yu Ma
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
166
|
Affiliation(s)
- Neil R Miller
- The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
167
|
Morris SM, Monroe CL, Gutmann DH. Macrocephaly Is Not a Predictor of Optic Pathway Glioma Development or Treatment in Neurofibromatosis Type 1. J Child Neurol 2016; 31:1540-1545. [PMID: 27581847 DOI: 10.1177/0883073816664659] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 07/08/2016] [Accepted: 07/22/2016] [Indexed: 12/26/2022]
Abstract
Neurofibromatosis type 1 is a common neurogenetic disorder characterized by significant clinical variability. As such, numerous studies have focused on identifying clinical, radiographic, or molecular biomarkers that predict the occurrence or progression of specific clinical features in individuals with neurofibromatosis type 1. One of these clinical biomarkers, macrocephaly, has been proposed as a prognostic factor for optic pathway glioma development. In the current study, the authors demonstrate that macrocephaly is not associated with the development of these brain tumors or the need to institute treatment for clinical progression. These findings suggest that macrocephaly is not a robust biomarker of optic pathway glioma formation or progression in children with neurofibromatosis type 1.
Collapse
Affiliation(s)
- Stephanie M Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Courtney L Monroe
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
168
|
Chiang JCH, Ellison DW. Molecular pathology of paediatric central nervous system tumours. J Pathol 2016; 241:159-172. [DOI: 10.1002/path.4813] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 09/23/2016] [Indexed: 12/26/2022]
Affiliation(s)
- Jason CH Chiang
- Department of Pathology; St Jude Children's Research Hospital; Memphis TN 38105 USA
| | - David W Ellison
- Department of Pathology; St Jude Children's Research Hospital; Memphis TN 38105 USA
| |
Collapse
|
169
|
Avery RA, Mansoor A, Idrees R, Trimboli-Heidler C, Ishikawa H, Packer RJ, Linguraru MG. Optic pathway glioma volume predicts retinal axon degeneration in neurofibromatosis type 1. Neurology 2016; 87:2403-2407. [PMID: 27815398 DOI: 10.1212/wnl.0000000000003402] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 08/19/2016] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVE To determine whether tumor size is associated with retinal nerve fiber layer (RNFL) thickness, a measure of axonal degeneration and an established biomarker of visual impairment in children with optic pathway gliomas (OPGs) secondary to neurofibromatosis type 1 (NF1). METHODS Children with NF1-OPGs involving the optic nerve (extension into the chiasm and tracts permitted) who underwent both volumetric MRI analysis and optical coherence tomography (OCT) within 2 weeks of each other were included. Volumetric measurement of the entire anterior visual pathway (AVP; optic nerve, chiasm, and tract) was performed using high-resolution T1-weighted MRI. OCT measured the average RNFL thickness around the optic nerve. Linear regression models evaluated the relationship between RNFL thickness and AVP dimensions and volume. RESULTS Thirty-eight participants contributed 55 study eyes. The mean age was 5.78 years. Twenty-two participants (58%) were female. RNFL thickness had a significant negative relationship to total AVP volume and total brain volume (p < 0.05, all comparisons). For every 1 mL increase in AVP volume, RNFL thickness declined by approximately 5 microns. A greater AVP volume of OPGs involving the optic nerve and chiasm, but not the tracts, was independently associated with a lower RNFL thickness (p < 0.05). All participants with an optic chiasm volume >1.3 mL demonstrated axonal damage (i.e., RNFL thickness <80 microns). CONCLUSIONS Greater OPG and AVP volume predicts axonal degeneration, a biomarker of vision loss, in children with NF1-OPGs. MRI volumetric measures may help stratify the risk of visual loss from NF1-OPGs.
Collapse
Affiliation(s)
- Robert A Avery
- From the Center for Neuroscience and Behavior (R.A.A., R.J.P.), The Gilbert Family Neurofibromatosis Institute (R.A.A., C.T.-H., R.J.P.), Sheikh Zayed Institute for Pediatric Surgical Innovation (A.M., M.G.L.), and The Brain Tumor Institute (R.J.P.), Children's National Health System; The George Washington University School of Medicine and Health Sciences (R.I., M.G.L.), Washington, DC; UPMC Eye Center, Eye and Ear Institute (H.I.), Ophthalmology and Visual Science Research Center, Department of Ophthalmology, University of Pittsburgh School of Medicine; and Department of Bioengineering (H.I.), Swanson School of Engineering, University of Pittsburgh, PA.
| | - Awais Mansoor
- From the Center for Neuroscience and Behavior (R.A.A., R.J.P.), The Gilbert Family Neurofibromatosis Institute (R.A.A., C.T.-H., R.J.P.), Sheikh Zayed Institute for Pediatric Surgical Innovation (A.M., M.G.L.), and The Brain Tumor Institute (R.J.P.), Children's National Health System; The George Washington University School of Medicine and Health Sciences (R.I., M.G.L.), Washington, DC; UPMC Eye Center, Eye and Ear Institute (H.I.), Ophthalmology and Visual Science Research Center, Department of Ophthalmology, University of Pittsburgh School of Medicine; and Department of Bioengineering (H.I.), Swanson School of Engineering, University of Pittsburgh, PA
| | - Rabia Idrees
- From the Center for Neuroscience and Behavior (R.A.A., R.J.P.), The Gilbert Family Neurofibromatosis Institute (R.A.A., C.T.-H., R.J.P.), Sheikh Zayed Institute for Pediatric Surgical Innovation (A.M., M.G.L.), and The Brain Tumor Institute (R.J.P.), Children's National Health System; The George Washington University School of Medicine and Health Sciences (R.I., M.G.L.), Washington, DC; UPMC Eye Center, Eye and Ear Institute (H.I.), Ophthalmology and Visual Science Research Center, Department of Ophthalmology, University of Pittsburgh School of Medicine; and Department of Bioengineering (H.I.), Swanson School of Engineering, University of Pittsburgh, PA
| | - Carmelina Trimboli-Heidler
- From the Center for Neuroscience and Behavior (R.A.A., R.J.P.), The Gilbert Family Neurofibromatosis Institute (R.A.A., C.T.-H., R.J.P.), Sheikh Zayed Institute for Pediatric Surgical Innovation (A.M., M.G.L.), and The Brain Tumor Institute (R.J.P.), Children's National Health System; The George Washington University School of Medicine and Health Sciences (R.I., M.G.L.), Washington, DC; UPMC Eye Center, Eye and Ear Institute (H.I.), Ophthalmology and Visual Science Research Center, Department of Ophthalmology, University of Pittsburgh School of Medicine; and Department of Bioengineering (H.I.), Swanson School of Engineering, University of Pittsburgh, PA
| | - Hiroshi Ishikawa
- From the Center for Neuroscience and Behavior (R.A.A., R.J.P.), The Gilbert Family Neurofibromatosis Institute (R.A.A., C.T.-H., R.J.P.), Sheikh Zayed Institute for Pediatric Surgical Innovation (A.M., M.G.L.), and The Brain Tumor Institute (R.J.P.), Children's National Health System; The George Washington University School of Medicine and Health Sciences (R.I., M.G.L.), Washington, DC; UPMC Eye Center, Eye and Ear Institute (H.I.), Ophthalmology and Visual Science Research Center, Department of Ophthalmology, University of Pittsburgh School of Medicine; and Department of Bioengineering (H.I.), Swanson School of Engineering, University of Pittsburgh, PA
| | - Roger J Packer
- From the Center for Neuroscience and Behavior (R.A.A., R.J.P.), The Gilbert Family Neurofibromatosis Institute (R.A.A., C.T.-H., R.J.P.), Sheikh Zayed Institute for Pediatric Surgical Innovation (A.M., M.G.L.), and The Brain Tumor Institute (R.J.P.), Children's National Health System; The George Washington University School of Medicine and Health Sciences (R.I., M.G.L.), Washington, DC; UPMC Eye Center, Eye and Ear Institute (H.I.), Ophthalmology and Visual Science Research Center, Department of Ophthalmology, University of Pittsburgh School of Medicine; and Department of Bioengineering (H.I.), Swanson School of Engineering, University of Pittsburgh, PA
| | - Marius George Linguraru
- From the Center for Neuroscience and Behavior (R.A.A., R.J.P.), The Gilbert Family Neurofibromatosis Institute (R.A.A., C.T.-H., R.J.P.), Sheikh Zayed Institute for Pediatric Surgical Innovation (A.M., M.G.L.), and The Brain Tumor Institute (R.J.P.), Children's National Health System; The George Washington University School of Medicine and Health Sciences (R.I., M.G.L.), Washington, DC; UPMC Eye Center, Eye and Ear Institute (H.I.), Ophthalmology and Visual Science Research Center, Department of Ophthalmology, University of Pittsburgh School of Medicine; and Department of Bioengineering (H.I.), Swanson School of Engineering, University of Pittsburgh, PA
| |
Collapse
|
170
|
Parkhurst E, Abboy S. Optic Gliomas in Neurofibromatosis Type 1. J Pediatr Ophthalmol Strabismus 2016; 53:334-338. [PMID: 27537249 DOI: 10.3928/01913913-20160810-03] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 06/10/2016] [Indexed: 11/20/2022]
Abstract
PURPOSE To examine the incidence, presentation, and outcome of optic gliomas in children with neurofibromatosis type 1 (NF1) in Southern California Kaiser Permanente. METHODS The authors queried the Southern California Kaiser Permanente electronic medical record database to find patients diagnosed as having NF1. Genetics, ophthalmology, and imaging medical records of patients with optic glioma were reviewed. RESULTS A total of 708 patients younger than 21 years had a diagnosis of NF1 in Southern California Kaiser Permanente and 30 (4.2%) had a diagnosis of optic glioma. The average age of diagnosis was 5 years, with a range of 18 months to 12 years. Half (15 of 30) of the patients diagnosed as having optic glioma presented with symptoms (eg, vision loss, proptosis, and precocious puberty). Eight of 15 of the symptomatic patients were treated with surgery and/or chemotherapy. Symptomatic children were diagnosed later than those diagnosed through routine screening (5.7 vs 3.9 years old). The oldest child presented with symptoms at age 12 years. One asymptomatic patient had prophylactic chemotherapy. Sixty-three percent (19 of 30) of the gliomas were bilateral, 23% (7 of 30) were right-sided, and 13% (4 of 30) were left-sided. Fifty-three percent (17 of 30) of the gliomas involved the optic chiasm. CONCLUSIONS Screening practices for optic glioma are inconsistent. Most children with NF1 at risk for optic glioma do not have even one visit with an ophthalmologist. Children with NF1 can develop asymptomatic optic glioma as early as age 1 year. Annual ophthalmologic examination and screening for precocious puberty in children with NF1 is important for early diagnosis of optic gliomas and may reduce morbidity. [J Pediatr Ophthalmol Strabismus. 2016;53(6):334-338.].
Collapse
|
171
|
Mikirova N, Hunnunghake R, Scimeca RC, Chinshaw C, Ali F, Brannon C, Riordan N. High-Dose Intravenous Vitamin C Treatment of a Child with Neurofibromatosis Type 1 and Optic Pathway Glioma: A Case Report. AMERICAN JOURNAL OF CASE REPORTS 2016; 17:774-781. [PMID: 27773919 PMCID: PMC5081233 DOI: 10.12659/ajcr.899754] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Patient: Male, 1 Final Diagnosis: Optic glioma Symptoms: Visual problems Medication: — Clinical Procedure: Intravenous vitamin C Specialty: Oncology
Collapse
Affiliation(s)
- Nina Mikirova
- Department of Research, Riordan Clinic, Wichita, KS, USA
| | | | - Ruth C Scimeca
- Department of Research, Riordan Clinic, Wichita, KS, USA
| | - Charles Chinshaw
- Department of Laboratory Analysis, Riordan Clinic, Wichita, KS, USA
| | - Faryal Ali
- Department of Laboratory Analysis, Riordan Clinic, Wichita, KS, USA
| | - Chris Brannon
- Department of Clinics, Riordan Clinic, Wichita, KS, USA
| | - Neil Riordan
- Department of Research, Inc., Riordan-McKenna Institute, Panama City, Panama
| |
Collapse
|
172
|
Abstract
The etiology of most childhood cancer remains largely unknown, but is likely attributable to random or induced genetic aberrations in somatic tissue. However, a subset of children develops cancer in the setting of an underlying inheritable condition involving a germline genetic mutation or chromosomal aberration. The term "neurocutaneous syndrome" encompasses a group of multisystem, hereditary disorders that are associated with skin manifestations as well as central and/or peripheral nervous system lesions of variable severity. This review outlines the central nervous system tumors associated with underlying neurocutaneous disorders, including neurofibromatosis type 1, neurofibromatosis type 2, schwannomatosis, tuberous sclerosis complex, Von Hippel Lindau, and nevoid basal cell carcinoma syndrome. Recognizing the presence of an underlying syndrome is critically important to both optimizing clinical care and treatment as well as genetic counseling and monitoring of these affected patients and their families.
Collapse
Affiliation(s)
- Nicole J Ullrich
- Department of Neurology, Boston Children's Hospital and Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
173
|
McCrea HJ, George E, Settler A, Schwartz TH, Greenfield JP. Pediatric Suprasellar Tumors. J Child Neurol 2016; 31:1367-76. [PMID: 26676303 DOI: 10.1177/0883073815620671] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 11/06/2015] [Indexed: 11/15/2022]
Abstract
The various childhood suprasellar tumors, while pathologically distinct, present similar clinical and surgical challenges as a result of their common anatomic location. These lesions are in close proximity to or may invade the optic nerve and chiasm, pituitary gland and infundibulum, hypothalamus, and third ventricle, leading to presenting features including visual field loss, impairment in visual acuity, endocrine dysfunction, and hydrocephalus. Though many suprasellar lesions are relatively benign in pathology, treatment may be complicated by high surgical morbidity resulting from damage to the hypothalamic-pituitary axis. Here we review the most frequent pediatric lesions occurring in the suprasellar region: craniopharyngioma, chiasmatic glioma, germ cell tumor, Rathke cleft and arachnoid cysts, pituitary adenoma, and histiocytosis. This review outlines both common presenting features and differentiating aspects of these lesions. It also includes classic radiographic presentations and treatment considerations for each lesion.
Collapse
Affiliation(s)
- Heather J McCrea
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY, USA
| | - Emilie George
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY, USA
| | - Allison Settler
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY, USA
| | - Theodore H Schwartz
- Departments of Neurological Surgery, Otolaryngology-Head and Neck Surgery, and Neurology and Neuroscience, Weill Cornell Medical College, New York, NY, USA
| | - Jeffrey P Greenfield
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
174
|
RNA Sequencing of Tumor-Associated Microglia Reveals Ccl5 as a Stromal Chemokine Critical for Neurofibromatosis-1 Glioma Growth. Neoplasia 2016; 17:776-88. [PMID: 26585233 PMCID: PMC4656811 DOI: 10.1016/j.neo.2015.10.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 10/06/2015] [Accepted: 10/14/2015] [Indexed: 12/31/2022] Open
Abstract
Solid cancers develop within a supportive microenvironment that promotes tumor formation and growth through the elaboration of mitogens and chemokines. Within these tumors, monocytes (macrophages and microglia) represent rich sources of these stromal factors. Leveraging a genetically engineered mouse model of neurofibromatosis type 1 (NF1) low-grade brain tumor (optic glioma), we have previously demonstrated that microglia are essential for glioma formation and maintenance. To identify potential tumor-associated microglial factors that support glioma growth (gliomagens), we initiated a comprehensive large-scale discovery effort using optimized RNA-sequencing methods focused specifically on glioma-associated microglia. Candidate microglial gliomagens were prioritized to identify potential secreted or membrane-bound proteins, which were next validated by quantitative real-time polymerase chain reaction as well as by RNA fluorescence in situ hybridization following minocycline-mediated microglial inactivation in vivo. Using these selection criteria, chemokine (C-C motif) ligand 5 (Ccl5) was identified as a chemokine highly expressed in genetically engineered Nf1 mouse optic gliomas relative to nonneoplastic optic nerves. As a candidate gliomagen, recombinant Ccl5 increased Nf1-deficient optic nerve astrocyte growth in vitro. Importantly, consistent with its critical role in maintaining tumor growth, treatment with Ccl5 neutralizing antibodies reduced Nf1 mouse optic glioma growth and improved retinal dysfunction in vivo. Collectively, these findings establish Ccl5 as an important microglial growth factor for low-grade glioma maintenance relevant to the development of future stroma-targeted brain tumor therapies.
Collapse
|
175
|
Mannino G, Abdolrahimzadeh B, Calafiore S, Anselmi G, Mannino C, Lambiase A. A review of the role of ultrasound biomicroscopy in glaucoma associated with rare diseases of the anterior segment. Clin Ophthalmol 2016; 10:1453-9. [PMID: 27536058 PMCID: PMC4975163 DOI: 10.2147/opth.s112166] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ultrasound biomicroscopy is a non-invasive imaging technique, which allows high-resolution evaluation of the anatomical features of the anterior segment of the eye regardless of optical media transparency. This technique provides diagnostically significant information in vivo for the cornea, anterior chamber, chamber angle, iris, posterior chamber, zonules, ciliary body, and lens, and is of great value in assessment of the mechanisms of glaucoma onset. The purpose of this paper is to review the use of ultrasound biomicroscopy in the diagnosis and management of rare diseases of the anterior segment such as mesodermal dysgenesis of the neural crest, iridocorneal endothelial syndrome, phakomatoses, and metabolic disorders.
Collapse
Affiliation(s)
- Giuseppe Mannino
- Ophthalmology Unit, Saint Andrea Hospital, Department of Neurosciences, Mental Health and Sense Organs, University of Rome "Sapienza", Rome, Italy
| | | | - Silvia Calafiore
- Ophthalmology Unit, Saint Andrea Hospital, Department of Neurosciences, Mental Health and Sense Organs, University of Rome "Sapienza", Rome, Italy
| | - Gianmario Anselmi
- Ophthalmology Unit, Saint Andrea Hospital, Department of Neurosciences, Mental Health and Sense Organs, University of Rome "Sapienza", Rome, Italy
| | - Cristina Mannino
- Ophthalmology Unit, Saint Andrea Hospital, Department of Neurosciences, Mental Health and Sense Organs, University of Rome "Sapienza", Rome, Italy
| | - Alessandro Lambiase
- Department of Sense Organs, Ophthalmology Unit, University of Rome "Sapienza", Rome, Italy
| |
Collapse
|
176
|
An Update on the Ophthalmologic Features in the Phakomatoses. J Ophthalmol 2016; 2016:3043026. [PMID: 27493794 PMCID: PMC4967465 DOI: 10.1155/2016/3043026] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 05/11/2016] [Accepted: 06/06/2016] [Indexed: 11/18/2022] Open
Abstract
Neurofibromatosis type 1, tuberous sclerosis complex, and Von Hippel-Lindau disease, historically classified as the phakomatoses, are hereditary multisystem disorders characterized by the presence of hamartoma, which carry the risk of malignant transformation. The alteration of tumor suppressor genes seems to be at the basis of their pathophysiogenetic mechanism. Lisch and choroidal nodules in neurofibromatosis type 1, retinal astrocytomas in tuberous sclerosis complex, and retinal capillary hemangioma in Von Hippel-Lindau disease are the principal ophthalmic hamartomatous manifestations. The advent of novel imaging techniques such as near infrared reflectance and optical coherence tomography has provided unprecedented insight on the choroidal and retinal features of these diseases. These methods have improved early diagnosis and the ongoing surveillance in these conditions. Among an array of treatment modalities, antivascular endothelial growth factor therapy has been used in the management of retinal hamartomas but results have been varied. This review is an update on the pathophysiogenetic mechanisms, ophthalmic manifestations, and novel treatment strategies in the phakomatoses with emphasis on the role of imaging techniques.
Collapse
|
177
|
Strabologische und neuroophthalmologische Aspekte kindlicher Tumorerkrankungen. Ophthalmologe 2016; 113:557-69. [DOI: 10.1007/s00347-016-0289-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
178
|
An Update on Neurofibromatosis Type 1: Not Just Café-au-Lait Spots and Freckling. Part II. Other Skin Manifestations Characteristic of NF1. NF1 and Cancer. ACTAS DERMO-SIFILIOGRAFICAS 2016. [DOI: 10.1016/j.adengl.2016.05.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
179
|
|
180
|
Helfferich J, Nijmeijer R, Brouwer OF, Boon M, Fock A, Hoving EW, Meijer L, den Dunnen WFA, de Bont ESJM. Neurofibromatosis type 1 associated low grade gliomas: A comparison with sporadic low grade gliomas. Crit Rev Oncol Hematol 2016; 104:30-41. [PMID: 27263935 DOI: 10.1016/j.critrevonc.2016.05.008] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 03/24/2016] [Accepted: 05/12/2016] [Indexed: 11/29/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant disorder, associated with a variable clinical phenotype including café-au-lait spots, intertriginous freckling, Lisch nodules, neurofibromas, optic pathway gliomas and distinctive bony lesions. NF1 is caused by a mutation in the NF1 gene, which codes for neurofibromin, a large protein involved in the MAPK- and the mTOR-pathway through RAS-RAF signalling. NF1 is a known tumour predisposition syndrome, associated with different tumours of the nervous system including low grade gliomas (LGGs) in the paediatric population. The focus of this review is on grade I pilocytic astrocytomas (PAs), the most commonly observed histologic subtype of low grade gliomas in NF1. Clinically, these PAs have a better prognosis and show different localisation patterns than their sporadic counterparts, which are most commonly associated with a KIAA1549:BRAF fusion. In this review, possible mechanisms of tumourigenesis in LGGs with and without NF1 will be discussed, including the contribution of different signalling pathways and tumour microenvironment. Furthermore we will discuss how increased understanding of tumourigenesis may lead to new potential targets for treatment.
Collapse
Affiliation(s)
- Jelte Helfferich
- Department of Paediatrics, Beatrix Children's Hospital, Paediatric Oncology/Hematology Division, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Neurology, Paediatric Neurology Division, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Ronald Nijmeijer
- Department of Pathology and Medical Biology, Pathology Division, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Oebele F Brouwer
- Department of Neurology, Paediatric Neurology Division, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Maartje Boon
- Department of Neurology, Paediatric Neurology Division, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Annemarie Fock
- Department of Neurology, Paediatric Neurology Division, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Eelco W Hoving
- Department of Neurosurgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Lisethe Meijer
- Department of Paediatrics, Beatrix Children's Hospital, Paediatric Oncology/Hematology Division, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Wilfred F A den Dunnen
- Department of Pathology and Medical Biology, Pathology Division, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Eveline S J M de Bont
- Department of Paediatrics, Beatrix Children's Hospital, Paediatric Oncology/Hematology Division, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
181
|
Abdolrahimzadeh B, Piraino DC, Albanese G, Cruciani F, Rahimi S. Neurofibromatosis: an update of ophthalmic characteristics and applications of optical coherence tomography. Clin Ophthalmol 2016; 10:851-60. [PMID: 27257370 PMCID: PMC4874640 DOI: 10.2147/opth.s102830] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Neurofibromatosis (NF) is a multisystem disorder and tumor predisposition syndrome caused by genetic mutation on chromosome 17-17q11.2 in NF type 1 (NF1), and on chromosome 22-22q12.2 in NF type 2. The disorder is characterized by considerable heterogeneity of clinical expression. NF1 is the form with the most characteristic ocular manifestations. Lisch nodules of the iris are among the well-known diagnostic criteria for the disease. Glaucoma and associated globe enlargement have been described in a significant proportion of patients with NF1 and orbital–facial involvement. Optic nerve glioma may cause strabismus and proptosis, and palpebral neurofibroma may reach considerable size and occasionally show malignant transformation. Near infrared reflectance has greatly contributed to enhancing our knowledge on choroidal alterations in NF1. Indeed, some authors have proposed to include these among the diagnostic criteria. Optical coherence tomography has given new insight on retinal alterations and is a noninvasive tool in the management of optic nerve gliomas in children. Ocular manifestations in NF type 2 can range from early-onset cataracts in up to 80% of cases to optic nerve hamartomas and combined pigment epithelial and retinal hamartomas.
Collapse
Affiliation(s)
| | - Domenica Carmen Piraino
- Section of Ophthalmology, Department of Sense Organs, University of Rome "Sapienza", Rome, Italy
| | - Giorgio Albanese
- Section of Ophthalmology, Department of Sense Organs, University of Rome "Sapienza", Rome, Italy
| | - Filippo Cruciani
- Section of Ophthalmology, Department of Sense Organs, University of Rome "Sapienza", Rome, Italy
| | - Siavash Rahimi
- Pathology Centre, Queen Alexandra Hospital, Portsmouth, UK
| |
Collapse
|
182
|
Avery RA, Mansoor A, Idrees R, Biggs E, Alsharid MA, Packer RJ, Linguraru MG. Quantitative MRI criteria for optic pathway enlargement in neurofibromatosis type 1. Neurology 2016; 86:2264-70. [PMID: 27170570 DOI: 10.1212/wnl.0000000000002771] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 03/17/2016] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE To determine quantitative size thresholds for enlargement of the optic nerve, chiasm, and tract in children with neurofibromatosis type 1 (NF1). METHODS Children 0.5-18.6 years of age who underwent high-resolution T1-weighted MRI were eligible for inclusion. This consisted of children with NF1 with or without optic pathway gliomas (OPGs) and a control group who did not have other acquired, systemic, or genetic conditions that could alter their anterior visual pathway (AVP). Maximum and average diameter and volume of AVP structures were calculated from reconstructed MRI images. Values above the 95th percentile from the controls were considered the threshold for defining an abnormally large AVP measure. RESULTS A total of 186 children (controls = 82; NF1noOPG = 54; NF1+OPG = 50) met inclusion criteria. NF1noOPG and NF1+OPG participants demonstrated greater maximum optic nerve diameter and volume, optic chiasm volume, and total brain volume compared to controls (p < 0.05, all comparisons). Total brain volume, rather than age, predicted optic nerve and chiasm volume in controls (p < 0.05). Applying the 95th percentile threshold to all NF1 participants, the maximum optic nerve diameter (3.9 mm) and AVP volumes resulted in few false-positive errors (specificity >80%, all comparisons). CONCLUSIONS Quantitative reference values for AVP enlargement will enhance the development of objective diagnostic criteria for OPGs secondary to NF1.
Collapse
Affiliation(s)
- Robert A Avery
- From the Center for Neuroscience and Behavior (R.A.A., R.J.P.), The Gilbert Family Neurofibromatosis Institute (R.A.A., R.J.P.), the Sheikh Zayed Institute for Pediatric Surgical Innovation (A.M., E.B., M.A.A., M.G.L.), and The Brain Tumor Institute (R.J.P.), Children's National Health System; The George Washington University (R.I.); and The George Washington University School of Medicine and Health Sciences (M.G.L.), Washington, DC.
| | - Awais Mansoor
- From the Center for Neuroscience and Behavior (R.A.A., R.J.P.), The Gilbert Family Neurofibromatosis Institute (R.A.A., R.J.P.), the Sheikh Zayed Institute for Pediatric Surgical Innovation (A.M., E.B., M.A.A., M.G.L.), and The Brain Tumor Institute (R.J.P.), Children's National Health System; The George Washington University (R.I.); and The George Washington University School of Medicine and Health Sciences (M.G.L.), Washington, DC
| | - Rabia Idrees
- From the Center for Neuroscience and Behavior (R.A.A., R.J.P.), The Gilbert Family Neurofibromatosis Institute (R.A.A., R.J.P.), the Sheikh Zayed Institute for Pediatric Surgical Innovation (A.M., E.B., M.A.A., M.G.L.), and The Brain Tumor Institute (R.J.P.), Children's National Health System; The George Washington University (R.I.); and The George Washington University School of Medicine and Health Sciences (M.G.L.), Washington, DC
| | - Elijah Biggs
- From the Center for Neuroscience and Behavior (R.A.A., R.J.P.), The Gilbert Family Neurofibromatosis Institute (R.A.A., R.J.P.), the Sheikh Zayed Institute for Pediatric Surgical Innovation (A.M., E.B., M.A.A., M.G.L.), and The Brain Tumor Institute (R.J.P.), Children's National Health System; The George Washington University (R.I.); and The George Washington University School of Medicine and Health Sciences (M.G.L.), Washington, DC
| | - Mohammad Ali Alsharid
- From the Center for Neuroscience and Behavior (R.A.A., R.J.P.), The Gilbert Family Neurofibromatosis Institute (R.A.A., R.J.P.), the Sheikh Zayed Institute for Pediatric Surgical Innovation (A.M., E.B., M.A.A., M.G.L.), and The Brain Tumor Institute (R.J.P.), Children's National Health System; The George Washington University (R.I.); and The George Washington University School of Medicine and Health Sciences (M.G.L.), Washington, DC
| | - Roger J Packer
- From the Center for Neuroscience and Behavior (R.A.A., R.J.P.), The Gilbert Family Neurofibromatosis Institute (R.A.A., R.J.P.), the Sheikh Zayed Institute for Pediatric Surgical Innovation (A.M., E.B., M.A.A., M.G.L.), and The Brain Tumor Institute (R.J.P.), Children's National Health System; The George Washington University (R.I.); and The George Washington University School of Medicine and Health Sciences (M.G.L.), Washington, DC
| | - Marius George Linguraru
- From the Center for Neuroscience and Behavior (R.A.A., R.J.P.), The Gilbert Family Neurofibromatosis Institute (R.A.A., R.J.P.), the Sheikh Zayed Institute for Pediatric Surgical Innovation (A.M., E.B., M.A.A., M.G.L.), and The Brain Tumor Institute (R.J.P.), Children's National Health System; The George Washington University (R.I.); and The George Washington University School of Medicine and Health Sciences (M.G.L.), Washington, DC
| |
Collapse
|
183
|
Augenbeteiligung bei Neurofibromatose. Ophthalmologe 2016; 113:443-52. [DOI: 10.1007/s00347-016-0237-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
184
|
Blakeley JO, Plotkin SR. Therapeutic advances for the tumors associated with neurofibromatosis type 1, type 2, and schwannomatosis. Neuro Oncol 2016; 18:624-38. [PMID: 26851632 PMCID: PMC4827037 DOI: 10.1093/neuonc/nov200] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 08/17/2015] [Indexed: 01/08/2023] Open
Abstract
Neurofibromatosis type 1 (NF1), neurofibromatosis type 2 (NF2), and schwannomatosis (SWN) are tumor-suppressor syndromes. Each syndrome is an orphan disease; however, the tumors that arise within them represent the most common tumors of the nervous system worldwide. Systematic investigation of the pathways impacted by the loss of function of neurofibromin (encoded byNF1) and merlin (encoded byNF2) have led to therapeutic advances for patients with NF1 and NF2. In the syndrome of SWN, the genetic landscape is more complex, with 2 known causative genes (SMARCB1andLZTR1) accounting for up to 50% of familial SWN patients. The understanding of the molecular underpinnings of these syndromes is developing rapidly and offers more therapeutic options for the patients. In addition, common sporadic cancers harbor somatic alterations inNF1(ie, glioblastoma, breast cancer, melanoma),NF2(ie, meningioma, mesothelioma) andSMARCB1(ie, atypical teratoid/rhabdoid tumors) such that advances in management of syndromic tumors may benefit patients both with and without germline mutations. In this review, we discuss the clinical and genetic features of NF1, NF2 and SWN, the therapeutic advances for the tumors that arise within these syndromes and the interaction between these rare tumor syndromes and the common tumors that share these mutations.
Collapse
Affiliation(s)
- Jaishri O Blakeley
- Neurology, Neurosurgery and Oncology, Johns Hopkins University, Baltimore, MD (J.O.B.); Neurology, Harvard Medical School, Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, MA (S.R.P.)
| | - Scott R Plotkin
- Neurology, Neurosurgery and Oncology, Johns Hopkins University, Baltimore, MD (J.O.B.); Neurology, Harvard Medical School, Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, MA (S.R.P.)
| |
Collapse
|
185
|
Ater JL, Xia C, Mazewski CM, Booth TN, Freyer DR, Packer RJ, Sposto R, Vezina G, Pollack IF. Nonrandomized comparison of neurofibromatosis type 1 and non-neurofibromatosis type 1 children who received carboplatin and vincristine for progressive low-grade glioma: A report from the Children's Oncology Group. Cancer 2016; 122:1928-36. [PMID: 27061921 DOI: 10.1002/cncr.29987] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/12/2016] [Accepted: 02/16/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND To evaluate tumor responses, event-free survival (EFS), overall survival (OS), and toxicity of chemotherapy, children with neurofibromatosis type 1 (NF1) and progressive low-grade glioma were enrolled into the Children's Oncology Group (COG) A9952 protocol and treated with carboplatin and vincristine (CV). METHODS Non-NF1 patients were randomized to CV or thioguanine, procarbazine, 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea, and vincristine in COG A9952. NF1 patients were assigned to CV only. NF1 patients and non-NF1 patients who were treated with CV were compared with respect to baseline characteristics, toxicity, tumor responses, EFS, and OS. RESULTS A total of 127 eligible patients with NF1 were nonrandomly assigned to CV: 42 NF1 patients (33%) had events, and 6 (4.7%) died. The 5-year EFS rate was 69% ± 4% for the CV-NF1 group and 39% ± 4% for the CV-non-NF1 group (P < .001). In a univariate analysis, NF1 children had a significantly higher tumor response rate and superior EFS and OS in comparison with CV-treated children without NF1. NF1 patients and non-NF1 patients differed significantly in amount of residual tumor, extent of resection, tumor location, and pathology. According to a multivariate analysis, NF1 was independently associated with better EFS (P < .001) but not with OS. NF1 patients also had a decreased risk of grade 3 or 4 toxicities in comparison with non-NF1 patients. Three second malignant neoplasms occurred in NF1 patients receiving CV (CV-NF1 group) at a median of 7.8 years (range, 7.3-9.4 years) after enrollment, but there were none in the non-NF1 group. CONCLUSIONS Children with NF1 tolerated CV well and had tumor response rates and EFS that were superior to those for children without NF1. Cancer 2016;122:1928-36. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Joann L Ater
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Caihong Xia
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Claire M Mazewski
- Children's Health Care Atlanta, Hematology-Oncology, Emory University School of Medicine, Atlanta, Georgia
| | - Timothy N Booth
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - David R Freyer
- Children's Hospital Los Angeles, Children's Center for Cancer and Blood Diseases, Los Angeles, California
| | - Roger J Packer
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Richard Sposto
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Gilbert Vezina
- Department of Neurology, Children's National Medical Center, Washington, DC
| | - Ian F Pollack
- Children's Hospital of Pittsburgh, Pediatric Neurosurgery, Pittsburgh, Pennsylvania
| |
Collapse
|
186
|
Dunning-Davies BM, Parker APJ. Annual review of children with neurofibromatosis type 1. Arch Dis Child Educ Pract Ed 2016; 101:102-11. [PMID: 26486853 DOI: 10.1136/archdischild-2014-308084] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 09/13/2015] [Indexed: 01/16/2023]
Abstract
We aim to provide a concise, evidence-based framework to assist secondary level, community and acute paediatricians during a 20-60 min annual review of children with neurofibromatosis type 1. This review does not cover all aspects of the disorder. We recognise the importance of an overview of the pathogenesis, molecular genetic testing, clinical manifestations and management; we shall cover some of this briefly, but this is not our focus here. We focus instead on the following areas: (A) what questions should be asked during annual review, (B) what should be included in a focused examination, (C) when to request further investigations and (D) when should a referral be made to tertiary specialists and other members of the multidisciplinary team. Ongoing debates regarding screening remain in certain areas, particularly regarding imaging and ophthalmology follow-up; here we summarise the differing opinions and make a recommendation based on the currently available evidence.
Collapse
Affiliation(s)
- B M Dunning-Davies
- Department of Paediatric Neuroscience, Addenbrooke's Hospital, Cambridge, UK
| | - A P J Parker
- Department of Paediatric Neuroscience, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
187
|
De novo development of gliomas in a child with neurofibromatosis type 1, fragile X and previously normal brain magnetic resonance imaging. Radiol Case Rep 2016; 11:33-5. [PMID: 26973730 PMCID: PMC4769616 DOI: 10.1016/j.radcr.2015.12.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 12/28/2015] [Accepted: 12/30/2015] [Indexed: 12/04/2022] Open
Abstract
Fifteen to 20% of children with neurofibromatosis type 1 develop low-grade glial neoplasms. However, since neuroimaging is not routinely obtained until a child is clinically symptomatic, little is known about presymptomatic radiographic characteristics of gliomas in this at-risk population. Herein, we describe a child with neurofibromatosis type 1 who initially had normal brain imaging before the development of multifocal gliomas. Comparison of these serial images demonstrated that brain tumors can arise de novo in children with this cancer predisposition syndrome, further underscoring the limited prognostic value of normal baseline magnetic resonance imaging.
Collapse
|
188
|
Hutter S, Piro RM, Waszak SM, Kehrer-Sawatzki H, Friedrich RE, Lassaletta A, Witt O, Korbel JO, Lichter P, Schuhmann MU, Pfister SM, Tabori U, Mautner VF, Jones DTW. No correlation between NF1 mutation position and risk of optic pathway glioma in 77 unrelated NF1 patients. Hum Genet 2016; 135:469-475. [PMID: 26969325 DOI: 10.1007/s00439-016-1646-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 02/06/2016] [Indexed: 01/08/2023]
Abstract
Neurofibromatosis type 1 (NF1) is a common monogenic disorder whereby affected individuals are predisposed to developing CNS tumors, including optic pathway gliomas (OPGs, occurring in ~15 to 20 % of cases). So far, no definite genotype-phenotype correlation determining NF1 patients at risk for tumor formation has been described, although enrichment for mutations in the 5' region of the NF1 gene in OPG patients has been suggested. We used whole exome sequencing, targeted sequencing, and copy number analysis to screen 77 unrelated NF1 patients with (n = 41) or without (n = 36; age ≥10 years) optic pathway glioma for germline NF1 alterations. We identified germline NF1 mutations in 69 of 77 patients (90 %), but no genotype-phenotype correlation was observed. Our data using a larger patient cohort did not confirm the previously reported clustering of mutations in the 5' region of the NF1 gene in patients with OPG. Thus, NF1 mutation location should not currently be used as a clinical criterion to assess the risk of developing OPGs.
Collapse
Affiliation(s)
- Sonja Hutter
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Rosario M Piro
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany.,Institute of Computer Science, Freie Universität Berlin, Berlin, Germany.,Institute of Medical Genetics and Human Genetics, Charité University Hospital, Berlin, Germany
| | - Sebastian M Waszak
- European Molecular Biology Laboratory (EMBL), Genome Biology Research Unit, Heidelberg, Germany
| | | | - Reinhard E Friedrich
- Department of Oral and Cranio-Maxillofacial Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Alvaro Lassaletta
- Division of Hematology/Oncology, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Olaf Witt
- German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Jan O Korbel
- European Molecular Biology Laboratory (EMBL), Genome Biology Research Unit, Heidelberg, Germany
| | - Peter Lichter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany
| | - Martin U Schuhmann
- Department of Neurosurgery, University Hospital Tübingen, Tübingen, Germany.,Centre for Neurofibromatosis, Centre for Rare Diseases Tübingen, Tübingen, Germany.,German Cancer Consortium (DKTK) Partner Site Tübingen, Tübingen, Germany
| | - Stefan M Pfister
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany.,Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Uri Tabori
- Division of Hematology/Oncology, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Victor F Mautner
- Department of Neurology, University Hospital Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| | - David T W Jones
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany. .,German Cancer Consortium (DKTK) Core Center Heidelberg, Heidelberg, Germany.
| |
Collapse
|
189
|
Hernández-Martín A, Duat-Rodríguez A. An Update on Neurofibromatosis Type 1: Not Just Café-au-Lait Spots and Freckling. Part II. Other Skin Manifestations Characteristic of NF1. NF1 and Cancer. ACTAS DERMO-SIFILIOGRAFICAS 2016; 107:465-73. [PMID: 26956402 DOI: 10.1016/j.ad.2016.01.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/07/2016] [Accepted: 01/17/2016] [Indexed: 12/16/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is the most common neurocutaneous syndrome and probably the one best known to dermatologists. Although the genetic locus of NF1 was identified on chromosome 17 in 1987, diagnosis of the disease is still based primarily on clinical observations. The 7 diagnostic criteria of the National Institutes of Health, which were established in 1988, include 3 skin manifestations (café-au-lait spots, freckling on flexural areas, and cutaneous neurofibromas). The age at which these diagnostic lesions appear is variable: onset can be late in some patients while others never develop certain symptoms. Definitive diagnosis may therefore be delayed by years. Although the appearance of the characteristic café-au-lait spots and freckling in the early years of childhood are very suggestive of the disease, these signs are not pathognomonic and, in isolation, do not constitute sufficient evidence to establish a definitive diagnosis. Thus, other diagnoses should be considered in patients whose only symptoms are café-au-lait spots and freckling. By contrast, the presence of multiple cutaneous neurofibromas or at least 1 plexiform neurofibroma is a very specific indication of NF1. Identification of the different types of neurofibroma allows us to confirm the diagnosis and initiate appropriate management.
Collapse
Affiliation(s)
- A Hernández-Martín
- Servicio de Dermatología Hospital Infantil del Niño Jesús. Madrid, España.
| | - A Duat-Rodríguez
- Servicio de Neurología, Hospital Infantil del Niño Jesús, Madrid, España
| |
Collapse
|
190
|
Doganis D, Pourtsidis A, Tsakiris K, Baka M, Kouri A, Bouhoutsou D, Varvoutsi M, Servitzoglou M, Dana H, Kosmidis H. Optic pathway glioma in children: 10 years of experience in a single institution. Pediatr Hematol Oncol 2016; 33:102-8. [PMID: 27007263 DOI: 10.3109/08880018.2016.1155101] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Optic pathway glioma (OPG) is a rare brain tumor that occurs more commonly during early childhood and is frequently associated with neurofibromatosis type 1 (NF1). In this study, our aim was to describe the characteristics, management, and outcome of patients with OPG. We retrospectively analyzed the clinical charts of all children diagnosed with OPG at our institution from 2003 to 2013. Twenty children (11 boys and 9 girls, median age: 5 and 3/12 years; NF1: 15/20) were diagnosed with OPG. The diagnosis was based on magnetic resonance imaging (MRI) findings. A biopsy was useful in 3 patients. The main reason for seeking medical advice was decreased vision (7/20 patients), whereas in 10/20 patients, the diagnosis was established during the routine follow-up for their NF1. Fifteen patients demonstrated MRI findings of optic nerve involvement and/or chiasmal tumor, whereas in 5 children, postchiasmal structures were also involved. Sixteen patients (16/20) received carboplatin-based regimens, whereas 4/20 patients were only under close observation. Six patients showed deterioration of visual acuity and/or imaging findings at the end of treatment and/or during their follow-up. Three of them (3/6) underwent tumor resection, whereas 1 (1/6) received radiation treatment. None of our patients had total blindness from both eyes. Half of our patients were diagnosed during follow-up for their NF1, the incidence of which was high in our group. Our data suggest that chemotherapy helps in the preservation of vision in the majority of children.
Collapse
Affiliation(s)
- Dimitrios Doganis
- a Department of Oncology , P. & A. Kyriakou Children's Hospital , Athens , Greece
| | - Apostolos Pourtsidis
- a Department of Oncology , P. & A. Kyriakou Children's Hospital , Athens , Greece
| | - Kleonikos Tsakiris
- b Department of Ophthalmology , P. & A. Kyriakou Children's Hospital , Athens , Greece
| | - Margarita Baka
- a Department of Oncology , P. & A. Kyriakou Children's Hospital , Athens , Greece
| | - Agathi Kouri
- b Department of Ophthalmology , P. & A. Kyriakou Children's Hospital , Athens , Greece
| | - Despina Bouhoutsou
- a Department of Oncology , P. & A. Kyriakou Children's Hospital , Athens , Greece
| | - Maria Varvoutsi
- a Department of Oncology , P. & A. Kyriakou Children's Hospital , Athens , Greece
| | - Marina Servitzoglou
- a Department of Oncology , P. & A. Kyriakou Children's Hospital , Athens , Greece
| | - Helen Dana
- a Department of Oncology , P. & A. Kyriakou Children's Hospital , Athens , Greece
| | - Helen Kosmidis
- a Department of Oncology , P. & A. Kyriakou Children's Hospital , Athens , Greece
| |
Collapse
|
191
|
Blanchard G, Lafforgue MP, Lion-François L, Kemlin I, Rodriguez D, Castelnau P, Carneiro M, Meyer P, Rivier F, Barbarot S, Chaix Y. Systematic MRI in NF1 children under six years of age for the diagnosis of optic pathway gliomas. Study and outcome of a French cohort. Eur J Paediatr Neurol 2016; 20:275-281. [PMID: 26774135 DOI: 10.1016/j.ejpn.2015.12.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 11/13/2015] [Accepted: 12/02/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND/PURPOSE Optic pathway glioma (OPG) is the most common central nervous system tumor in children with neurofibromatosis type 1 (NF1), affecting 15-20% of patients. We reviewed the medical records of children systematically screened by ophthalmologic and MRI examinations to determine the influence of screening on the therapeutic management of children with OPG. METHODS Data were collected on 306 newly diagnosed cases screened with systematic MRI from January 2001 to July 2007. In the OPG group, we distinguished the asymptomatic or symptomatic groups according to their initial status. RESULTS Forty-five patients had confirmed OPG (14.7%). Thirty-six patients (80%) were asymptomatic and nine (20%) were symptomatic at the time of diagnosis with visual symptoms in six cases. The average age at OPG diagnosis was 3.4 years with six patients (13%) over six years old. Average follow-up was 7.7 years. Progression was observed in 16 cases (35%). Most patient conditions were managed conservatively (87%). Six children (13%) were treated with chemotherapy due to worsening visual function. All of these children had severe or mild visual impairment at the end of follow-up. CONCLUSION Our study does not support a clear benefit of systematic MRI screening in NF1 children under six years old. Systematic neuroimaging in our study did not influence therapeutic management. Although OPG diagnosis was made early, treatment with chemotherapy did not improve the final visual outcome. If MRI remains the best tool for the diagnosis of cerebral and spinal pathologies in the NF1 population, our current study questions the usefulness of systematic MRI screening for OPG diagnosis. Conversely, this study suggests that the indication of neuroimaging should be dictated by the results of annual clinical and ophthalmological assessments.
Collapse
Affiliation(s)
- Gaelle Blanchard
- Service de neurologie pédiatrique, Hôpital Femme, Mère, Enfant, Bron, France
| | | | | | - Isabelle Kemlin
- AP-HP, Service de Neurologie Pédiatrique et Centre de Référence des Neurofibromatoses, Hôpital Armand Trousseau, CHU Paris Est, Paris, France
| | - Diana Rodriguez
- AP-HP, Service de Neurologie Pédiatrique et Centre de Référence des Neurofibromatoses, Hôpital Armand Trousseau, CHU Paris Est, Paris, France; Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Inserm U1141, Paris, France
| | - Pierre Castelnau
- Unité de Neuropédiatrie et Handicaps, Hôpital d'Enfants Gatien de Clocheville, CHU de Tours, France; Inserm, Imagerie et Cerveau, UMR 930, CHRU de Tours, Hôpital Bretonneau, Tours, France; Université de Tours, François Rabelais, Tours, France
| | - Maryline Carneiro
- CHRU de Montpellier, Neuropédiatrie & Centre de Référence des Troubles du Langage, Montpellier, France
| | - Pierre Meyer
- CHRU de Montpellier, Neuropédiatrie & Centre de Référence des Troubles du Langage, Montpellier, France
| | - François Rivier
- CHRU de Montpellier, Neuropédiatrie & Centre de Référence des Troubles du Langage, Montpellier, France; Université de Montpellier, UFR de Médecine, Montpellier, France; U1046 Inserm, UMR9214 CNRS, PhyMedExp, Université de Montpellier, Montpellier, France
| | | | - Yves Chaix
- Hôpital des Enfants, CHU Purpan, Toulouse, France; Inserm, Imagerie Cérébrale et Handicaps Neurologiques UMR 825, CHU Purpan, Toulouse, France; Université de Toulouse III - Paul Sabatier, Toulouse, France.
| | | |
Collapse
|
192
|
Toonen JA, Anastasaki C, Smithson LJ, Gianino SM, Li K, Kesterson RA, Gutmann DH. NF1 germline mutation differentially dictates optic glioma formation and growth in neurofibromatosis-1. Hum Mol Genet 2016; 25:1703-13. [PMID: 26908603 PMCID: PMC4986327 DOI: 10.1093/hmg/ddw039] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 02/08/2016] [Indexed: 12/24/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is a common neurogenetic condition characterized by significant clinical heterogeneity. A major barrier to developing precision medicine approaches for NF1 is an incomplete understanding of the factors that underlie its inherent variability. To determine the impact of the germline NF1 gene mutation on the optic gliomas frequently encountered in children with NF1, we developed genetically engineered mice harboring two representative NF1-patient-derived Nf1 gene mutations (c.2542G>C;p.G848R and c.2041C>T;p.R681X). We found that each germline Nf1 gene mutation resulted in different levels of neurofibromin expression. Importantly, only R681X(CKO) but not G848R(CKO), mice develop optic gliomas with increased optic nerve volumes, glial fibrillary acid protein immunoreactivity, proliferation and retinal ganglion cell death, similar to Nf1 conditional knockout mice harboring a neomycin insertion (neo) as the germline Nf1 gene mutation. These differences in optic glioma phenotypes reflect both cell-autonomous and stromal effects of the germline Nf1 gene mutation. In this regard, primary astrocytes harboring the R681X germline Nf1 gene mutation exhibit increased basal astrocyte proliferation (BrdU incorporation) indistinguishable from neo(CKO) astrocytes, whereas astrocytes with the G848R mutation have lower levels of proliferation. Evidence for paracrine effects from the tumor microenvironment were revealed when R681X(CKO) mice were compared with conventional neo(CKO) mice. Relative to neo(CKO) mice, the optic gliomas from R681X(CKO) mice had more microglia infiltration and JNK(Thr183/Tyr185) activation, microglia-produced Ccl5, and glial AKT(Thr308) activation. Collectively, these studies establish that the germline Nf1 gene mutation is a major determinant of optic glioma development and growth through by both tumor cell-intrinsic and stromal effects.
Collapse
Affiliation(s)
- Joseph A Toonen
- Department of Neurology, Washington University School of Medicine, PO Box 8111, 660 S. Euclid Avenue, St. Louis, MO 63110, USA and
| | - Corina Anastasaki
- Department of Neurology, Washington University School of Medicine, PO Box 8111, 660 S. Euclid Avenue, St. Louis, MO 63110, USA and
| | - Laura J Smithson
- Department of Neurology, Washington University School of Medicine, PO Box 8111, 660 S. Euclid Avenue, St. Louis, MO 63110, USA and
| | - Scott M Gianino
- Department of Neurology, Washington University School of Medicine, PO Box 8111, 660 S. Euclid Avenue, St. Louis, MO 63110, USA and
| | - Kairong Li
- Department of Genetics, University of Alabama, Birmingham, AL 35233, USA
| | - Robert A Kesterson
- Department of Genetics, University of Alabama, Birmingham, AL 35233, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, PO Box 8111, 660 S. Euclid Avenue, St. Louis, MO 63110, USA and
| |
Collapse
|
193
|
Bakker AC, La Rosa S, Sherman LS, Knight P, Lee H, Pancza P, Nievo M. Neurofibromatosis as a gateway to better treatment for a variety of malignancies. Prog Neurobiol 2016; 152:149-165. [PMID: 26854064 DOI: 10.1016/j.pneurobio.2016.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 01/25/2016] [Accepted: 01/25/2016] [Indexed: 12/23/2022]
Abstract
The neurofibromatoses (NF) are a group of rare genetic disorders that can affect all races equally at an incidence from 1:3000 (NF1) to a log unit lower for NF2 and schwannomatosis. Since the research community is reporting an increasing number of malignant cancers that carry mutations in the NF genes, the general interest of both the research and pharma community is increasing and the authors saw an opportunity to present a novel, fresh approach to drug discovery in NF. The aim of the paper is to challenge the current drug discovery approach to NF, whereby existing targeted therapies that are either in the clinic or on the market for other disease indications are repurposed for NF. We offer a suggestion for an alternative drug discovery approach. In the new approach, selective and tolerable targeted therapies would be developed for NF and later expanded to patients with more complex diseases such as malignant cancer in which the NF downstream pathways are deregulated. The Children's Tumor Foundation, together with some other major NF funders, is playing a key role in funding critical initiatives that will accelerate the development of better targeted therapies for NF patients, while these novel, innovative treatments could potentially be beneficial to molecularly characterized cancer patients in which NF mutations have been identified.
Collapse
Affiliation(s)
- Annette C Bakker
- Children's Tumor Foundation, 120, Wall Street, 16th Floor, New York 10005, United States
| | - Salvatore La Rosa
- Children's Tumor Foundation, 120, Wall Street, 16th Floor, New York 10005, United States
| | - Larry S Sherman
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR 97006, United States
| | - Pamela Knight
- Children's Tumor Foundation, 120, Wall Street, 16th Floor, New York 10005, United States
| | - Hyerim Lee
- Children's Tumor Foundation, 120, Wall Street, 16th Floor, New York 10005, United States
| | - Patrice Pancza
- Children's Tumor Foundation, 120, Wall Street, 16th Floor, New York 10005, United States
| | - Marco Nievo
- Children's Tumor Foundation, 120, Wall Street, 16th Floor, New York 10005, United States.
| |
Collapse
|
194
|
de Blank PMK, Fisher MJ, Lu L, Leisenring WM, Ness KK, Sklar CA, Stovall M, Vukadinovich C, Robison LL, Armstrong GT, Krull KR. Impact of vision loss among survivors of childhood central nervous system astroglial tumors. Cancer 2016; 122:730-9. [PMID: 26755438 DOI: 10.1002/cncr.29705] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 04/23/2015] [Accepted: 04/24/2015] [Indexed: 11/09/2022]
Abstract
BACKGROUND The impact of impaired vision on cognitive and psychosocial outcomes among long-term survivors of childhood low-grade gliomas has not been investigated previously but could inform therapeutic decision making. METHODS Data from the Childhood Cancer Survivor Study were used to investigate psychological outcomes (measures of cognitive/emotional function) and socioeconomic outcomes (education, income, employment, marital status, and independent living) among astroglial tumor survivors grouped by 1) vision without impairment, 2) vision with impairment (including unilateral blindness, visual field deficits, and amblyopia), or 3) bilateral blindness. The effect of vision status on outcomes was examined with multivariate logistic regression with adjustments for age, sex, cranial radiation therapy, and medical comorbidities. RESULTS Among 1233 survivors of childhood astroglial tumors 5 or more years after their diagnosis, 277 (22.5%) had visual impairment. In a multivariate analysis, survivors with bilateral blindness were more likely to be unmarried (adjusted odds ratio (OR), 4.7; 95% confidence interval [CI], 1.5-15.0), live with a caregiver (adjusted OR, 3.1; 95% CI, 1.3-7.5), and be unemployed (adjusted OR, 2.2; 95% CI, 1.1-4.5) in comparison with those without visual impairment. Bilateral blindness had no measurable effect on cognitive or emotional outcomes, and vision with impairment was not significantly associated with any psychological or socioeconomic outcomes. CONCLUSIONS Adult survivors of childhood astroglial tumors with bilateral blindness were more likely to live unmarried and dependently and to be unemployed. Survivors with visual impairment but some remaining vision did not differ significantly with respect to psychological function and socioeconomic status from those without visual impairment. Cancer 2016;122:730-739. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Peter M K de Blank
- Division of Pediatric Hematology and Oncology, Rainbow Babies & Children's Hospital, Cleveland, Ohio.,Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - Michael J Fisher
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lu Lu
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Wendy M Leisenring
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Kirsten K Ness
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Charles A Sklar
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center/Weill Cornell Medical College, New York, New York
| | - Marilyn Stovall
- Division of Radiation Oncology, Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chris Vukadinovich
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Leslie L Robison
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Gregory T Armstrong
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Kevin R Krull
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
195
|
SPECTRAL DOMAIN OPTICAL COHERENCE TOMOGRAPHY EVIDENCE OF RETINAL NERVE FIBER LAYER AND GANGLION CELL LOSS IN ADULT PATIENTS WITH NEUROFIBROMATOSIS TYPE 1. Retina 2016; 36:75-81. [DOI: 10.1097/iae.0000000000000650] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
196
|
Abstract
Pilocytic astrocytoma (PA) is the most common pediatric brain tumor in children. PAs are a distinct histologic and biologic subset of glioma that have a slow growth rate and may even spontaneously regress. These tumors tend to arise in the cerebellum and chiasmatic/hypothalamic region, but can also occur in other regions of the central nervous system. Dissemination is uncommon, but may occur in newly diagnosed PAs. Alterations in the Ras/RAF/mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway (Ras/ERK) have been discovered in a majority of PAs, with KIAA1549-BRAF fusions being the most commonly identified alteration. Children with neurofibromatosis 1 are predisposed to developing PAs, primarily within the optic pathway. When required, treatment consists of surgery, chemotherapy, and/or radiation, although new molecular agents targeting the Ras/ERK and related signaling pathways are promising new approaches. The 10-year survival rates are greater than 90% in pediatric patients; however, they are poorer in adults. Tumors that are amenable to complete resection (i.e., cerebellum and cortex) have the best overall survival.
Collapse
Affiliation(s)
- Miriam Bornhorst
- Gilbert Family Neurofibromatosis Institute and Brain Tumor Institute, Children's National Health System, Washington, DC, USA
| | - Didier Frappaz
- Department of Pediatric and Adult Neuro-oncology, Centre Léon Bérard and Institute of Pediatric Hematology and Oncology, Lyon, France
| | - Roger J Packer
- Gilbert Family Neurofibromatosis Institute and Brain Tumor Institute, Children's National Health System, Washington, DC, USA.
| |
Collapse
|
197
|
Abstract
Oculocutaneous albinism, Menkes syndrome, tuberous sclerosis, neurofibromatosis type 1, dyskeratosis congenita, lentiginosis profusa syndrome, incontinentia pigmenti, and Waardenburg syndrome all are genodermatoses that have well established gene mutations affecting multiple biological pathways, including melanin synthesis, copper transport, cellular proliferation, telomerase function, apoptosis, and melanocyte biology. Onchocerciasis results from a systemic inflammatory response to a nematode infection. Hypomelanosis of Ito is caused by chromosomal mosaicism, which underlies its phenotypic heterogeneity. Incomplete migration of melanocytes to the epidermis and other organs is the underlying feature of nevus of Ota. Vogt-Koyangi-Harada and vitiligo have an autoimmune etiology; the former is associated with considerable multiorgan involvement, while the latter is predominantly skin-limited.
Collapse
Affiliation(s)
- Syril Keena T Que
- Department of Dermatology, University of Connecticut Health Center, 21 South Road, Farmington, CT 06032
| | - Gillian Weston
- Department of Dermatology, University of Connecticut Health Center, 21 South Road, Farmington, CT 06032
| | - Jeanine Suchecki
- Department of Ophthalmology, University of Connecticut Health Center, 21 South Road, Farmington, CT 06032
| | - Janelle Ricketts
- Department of Dermatology, University of Connecticut Health Center, 21 South Road, Farmington, CT 06032.
| |
Collapse
|
198
|
Friedrich RE, Hagel C, Mautner VF. Unilateral gynaecomastia in a 16-month-old boy with neurofibromatosis type 1 - case report and brief review of the literature. GMS INTERDISCIPLINARY PLASTIC AND RECONSTRUCTIVE SURGERY DGPW 2015; 4:Doc11. [PMID: 26668786 PMCID: PMC4670967 DOI: 10.3205/iprs000070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant disease that shows high penetrance with a wide variability in the phenotype. Prepubertal enlargement of the breast in male subjects affected by this condition is well known, but rarely reported. The present case report describes diagnosis and therapy of unilateral gynaecomastia in a toddler showing integumental stigmata of NF1. Furthermore, the report provides a brief review of the literature concerning this finding in NF1. According to this review, the present case appears to be one of the youngest NF1-affected males affected by gynaecomastia that has been reported.
Collapse
Affiliation(s)
- Reinhard E Friedrich
- Department of Oral and Craniomaxillofacial Surgery, Eppendorf University Hospital, University of Hamburg, Hamburg, Germany
| | - Christian Hagel
- Institute of Neuropathology, Eppendorf University Hospital, University of Hamburg, Hamburg, Germany
| | - Victor-Felix Mautner
- Department of Neurology, Eppendorf University Hospital, University of Hamburg, Hamburg, Germany
| |
Collapse
|
199
|
Abstract
Neurocutaneous syndromes are a heterogeneous group of congenital and hereditary disorders with manifestations in the skin and the nervous system, usually together with ocular features that represent diagnostic clues and potential sources of morbidity. Dermatologists and ophthalmologists often need to work together in identifying and managing patients with these conditions; herein, we focus on classic and under-recognized neurocutaneous syndromes. We begin with autosomal dominant genodermatoses characterized by hamartomas and tumors in the skin, eyes, and central nervous system: neurofibromatosis type 1, tuberous sclerosis complex, and PTEN hamartoma-tumor syndrome. This is followed by a discussion of two mosaic disorders, Sturge-Weber syndrome and neurocutaneous melanocytosis. In addition to providing an update on clinical presentations and evaluation of patients with these conditions, we review recent insights into their pathogenesis, drawing attention to relationships among the diseases on a molecular level and implications regarding treatment. We also highlight the major features of other neurocutaneous syndromes that have ocular findings plus pigmentary, vascular, hyperkeratotic, adnexal, connective tissue, photosensitive, and inflammatory manifestations in the skin.
Collapse
Affiliation(s)
- Karen A Chernoff
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY 10016, USA
| | - Julie V Schaffer
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
200
|
Acquired nystagmus as the initial presenting sign of chiasmal glioma in young children. Eur J Paediatr Neurol 2015; 19:694-700. [PMID: 26190013 DOI: 10.1016/j.ejpn.2015.06.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 05/21/2015] [Accepted: 06/22/2015] [Indexed: 11/21/2022]
Abstract
BACKGROUND/PURPOSE The aim of the study was to investigate the incidence of nystagmus at diagnosis in children with optic pathway glioma involving the chiasm and hypothalamus. METHODS Twenty-two patients with a measurable optic pathway/hypothalamic glioma (without neurofibromatosis-1) were followed in our center from 2001 to 2013. The medical files were retrospectively reviewed for demographic and clinical findings, and the imaging scans, for tumor characteristics. RESULTS There were 9 boys and 13 girls of mean age 3.5 ± 4.4 years at diagnosis; 15 were aged <2 years. Tumor size ranged from 10 × 6 mm to 62 × 29 mm. Mean duration of follow-up was 8.3 ± 5.4 years. Nystagmus was detected at diagnosis in 10 children (45%), all <2 years old (66.6% of the younger group); no child older than 2 years presented with nystagmus. Nystagmus, once present, did not resolve and continued throughout follow-up. There were no cases of new onset of nystagmus during follow-up in the children in whom it was not detected at diagnosis. Treatment consisted of partial resection/biopsy with/without shunting (n = 13) and chemotherapy (n = 19) with (n = 2) or without adjuvant radiation. Of the 22 children, 6 had a radiographic response to treatment, 8 remained stable, and 8 (all of whom received chemotherapy) showed disease progression despite treatment. CONCLUSION In conclusion, monocular nystagmus is a more common presenting sign of optic pathway/hypothalamic glioma in children <2 years old than previously estimated. Although subtle, nystagmus has a very narrow differential diagnosis, and its presence should raise suspicions of a chiasmal tumor with prompt referral for imaging. The visual prognosis is moderate to poor.
Collapse
|