1
|
Soleman J, Constantini S, Roth J. Incidental brain tumor findings in children: prevalence, natural history, management, controversies, challenges, and dilemmas. Childs Nerv Syst 2024; 40:3179-3187. [PMID: 39215810 PMCID: PMC11511734 DOI: 10.1007/s00381-024-06598-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Incidental brain tumor findings in children involve the unexpected discovery of brain lesions during imaging for unrelated reasons. These findings differ significantly from those in adults, requiring a focus on pediatric-specific approaches in neurosurgery, neuroimaging, and neuro-oncology. Understanding the prevalence, progression, and management of these incidentalomas is crucial for informed decision-making, balancing patient welfare with the risks and benefits of intervention. Incidental brain tumors are observed in about 0.04-5.7% of cases, with most suspected low-grade lesions in children showing a benign course, though up to 3% may undergo malignant transformation. Treatment decisions are influenced by factors such as patient age, tumor characteristics, and family anxiety, with conservative management through surveillance often preferred. However, upfront surgery may be considered in cases with low surgical risk. Initial follow-up typically involves a comprehensive MRI after three months, with subsequent scans spaced out if the lesion remains stable. Changes in imaging or symptoms during follow-up could indicate malignant transformation, prompting consideration of surgery or biopsy. Several challenges and controversies persist, including the role of upfront biopsy for molecular profiling, the use of advanced imaging techniques like PET-CT and magnetic resonance spectroscopy, and the implications of the child's age at diagnosis. These issues highlight the need for further research to guide management and improve outcomes in pediatric patients with incidental brain tumor findings.
Collapse
Affiliation(s)
- Jehuda Soleman
- Department of Neurosurgery and Pediatric Neurosurgery, University Hospital and Children's Hospital Basel, Spitalstrasse 21, Basel, 4031, Switzerland.
- Faculty of Medicine, University of Basel, Basel, Switzerland.
| | - Shlomi Constantini
- Department of Pediatric Neurosurgery, Dana Children's Hospital, Tel Aviv Medical Center, Tel Aviv-Yafo, Israel
| | - Jonathan Roth
- Department of Pediatric Neurosurgery, Dana Children's Hospital, Tel Aviv Medical Center, Tel Aviv-Yafo, Israel
| |
Collapse
|
2
|
Albalkhi I, Shafqat A, Bin-Alamer O, Mallela AN, Gersey ZC, Fernandes Cabral D, Sabbagh AJ, Hadjipanayis CG, González-Martínez JA, Friedlander RM, Abou-Al-Shaar H. Complications and visual outcomes following surgical resection of pediatric optic pathway/hypothalamic gliomas: a systematic review and meta-analysis. Childs Nerv Syst 2024; 40:2033-2042. [PMID: 38649470 DOI: 10.1007/s00381-024-06407-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024]
Abstract
Pediatric optic pathway/hypothalamic gliomas (OPHG) pose challenges in treatment due to their location and proximity to vital structures. Surgical resection plays a key role in the management of OPHG especially when the tumor exhibits mass effect and causes symptoms. However, data regarding outcomes and complications of surgical resection for OPHG remains heterogenous. The authors performed a systematic review on pediatric OPHG in four databases: PubMed, EMBASE, Cochrane Library, and Google Scholar. We included studies that reported on the visual outcomes and complications of OPHG resection. A meta-analysis was performed and reported per the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. A total of 26 retrospective studies were included. Seven hundred ninety-seven pediatric patients with OPHG undergoing surgical resection were examined. A diagnosis of NF1 was confirmed in 9.7%. Gross total resection was achieved in 36.7%. Intraorbital optic pathway gliomas showed a significantly higher gross total resection rate compared to those located in the chiasmatic/hypothalamic region (75.8% vs. 9.6%). Postoperatively, visual acuity improved in 24.6%, remained unchanged in 68.2%, and worsened in 18.2%. Complications included hydrocephalus (35.4%), anterior pituitary dysfunction (19.6%), and transient diabetes insipidus (29%). Tumor progression post-resection occurred in 12.8%, through a mean follow-up of 53.5 months. Surgical resection remains an essential strategy for treating symptomatic and large pediatric OPHG and can result in favorable vision outcomes in most patients. Careful patient selection is critical. Patients should be monitored for hydrocephalus development postoperatively and followed up to assess for tumor progression and adjuvant treatment necessity.
Collapse
Affiliation(s)
- Ibrahem Albalkhi
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Department of Neuroradiology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Othman Bin-Alamer
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Arka N Mallela
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Zachary C Gersey
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - David Fernandes Cabral
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Abdulrahman J Sabbagh
- Department of Surgery, Division of Neurosurgery, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | | | - Robert M Friedlander
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Hussam Abou-Al-Shaar
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Fangusaro J, Avery RA, Fisher MJ, Packer RJ, Walsh KS, Schouten-van Meeteren A, Karres D, Bradford D, Bhatnagar V, Singh H, Kluetz PG, Donoghue M, Duke ES. Considering Functional Outcomes as Efficacy Endpoints in Pediatric Low-Grade Glioma Clinical Trials: An FDA Educational Symposium. Clin Cancer Res 2024; 30:2303-2308. [PMID: 38358393 PMCID: PMC11147731 DOI: 10.1158/1078-0432.ccr-23-3386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/22/2023] [Accepted: 02/13/2024] [Indexed: 02/16/2024]
Abstract
In October 2022, the FDA Oncology Center of Excellence hosted an educational symposium entitled, "Considering Functional Outcomes as Efficacy Endpoints in Pediatric Low-Grade Glioma (pLGG) Clinical Trials." The symposium brought together patient advocates, regulators from the FDA and the European Medicines Agency (EMA), and an international group of academic thought leaders in the field of pediatric neuro-oncology to discuss the potential role of functional outcomes, including visual acuity, motor function, and neurocognitive performance, as endpoints in clinical trials enrolling patients with pLGG. The panel discussed challenges and opportunities regarding the selection, implementation, and evaluation of clinical outcome assessments in these functional domains and outlined key considerations for their inclusion in future clinical trial design and role in new drug development.
Collapse
Affiliation(s)
- Jason Fangusaro
- Children's Hospital of Atlanta, Emory University and the Aflac Cancer Center, Atlanta, Georgia
| | - Robert A Avery
- The Children's Hospital of Philadelphia and the University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Michael J Fisher
- The Children's Hospital of Philadelphia and the University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Roger J Packer
- Center for Neuroscience and Behavioral Medicine and Brain Tumor Institute, Children's National Hospital, and The George Washington University School of Medicine, Washington, DC
| | - Karin S Walsh
- Center for Neuroscience and Behavioral Medicine and Brain Tumor Institute, Children's National Hospital, and The George Washington University School of Medicine, Washington, DC
| | | | - Dominik Karres
- Pediatric Medicines Office, Scientific Evidence Generation Department, Human Medicines Division, European Medicines Agency (EMA), Amsterdam, the Netherlands
| | - Diana Bradford
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Vishal Bhatnagar
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Harpreet Singh
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Paul G Kluetz
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Martha Donoghue
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Elizabeth S Duke
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
4
|
Bennebroek CA, Schouten CR, Montauban-van Swijndregt MC, Saeed P, Porro GL, Pott JWR, Dittrich ATM, Oostenbrink R, Schouten-van Meeteren AY, de Jong MC, de Graaf P. Treatment evaluation by volumetric segmentation in pediatric optic pathway glioma: evaluation of the effect of bevacizumab on intra-tumor components. J Neurooncol 2024; 166:79-87. [PMID: 38150061 DOI: 10.1007/s11060-023-04516-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/16/2023] [Indexed: 12/28/2023]
Abstract
PURPOSE Progressive pediatric optic pathway gliomas (OPGs) are treated by diverse systemic antitumor modalities. Refined insights on the course of intra-tumoral components are limited. METHODS We performed an exploratory study on the longitudinal volumetric course of different (intra-)tumor components by manual segmentation of MRI at the start and after 3, 6 and 12 months of bevacizumab (BVZ) treatment. RESULTS Thirty-one patients were treated with BVZ (median 12 months, range: 2-39 months). During treatment the total tumor volume decreased with median 19.9% (range: - 62.3 to + 29.7%; n = 30) within the first 3 months, decreased 19.0% (range: - 68.8 to + 96.1%; n = 28) between start and 6 months and 27.2% (range: -73.4 to + 36.0%; n = 21) between start and 12 months. Intra-tumoral cysts were present in 12 OPGs, all showed a decrease of volume during treatment. The relative contrast enhanced volume of NF1 associated OPG (n = 11) showed an significant reduction compared to OPG with a KIAA1549-BRAF fusion (p < 0.01). Three OPGs progressed during treatment, but were not preceded by an increase of relative contrast enhancement. CONCLUSION Treatment with BVZ of progressive pediatric OPGs leads to a decrease of both total tumor volume and cystic volume for the majority of OPGs with emphasis on the first three months. NF1 and KIAA1549-BRAF fusion related OPGs showed a different (early) treatment effect regarding the tumor enhancing component on MRI, which did not correlate with tumor volume changes. Future research is necessary to further evaluate these findings and its relevance to clinical outcome parameters.
Collapse
Affiliation(s)
- Carlien A Bennebroek
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1100DD, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, The Netherlands.
| | - Christiaan R Schouten
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Peerooz Saeed
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1100DD, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, The Netherlands
| | - Giorgio L Porro
- Department of Ophthalmology, Utrecht University Medical Center, Utrecht, The Netherlands
| | - Jan W R Pott
- Department of Ophthalmology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Anne T M Dittrich
- Department of Pediatrics, Radboud University Medical Center, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Rianne Oostenbrink
- ENCORE-NF1 Center, Department of General Pediatrics, Erasmus MC, Rotterdam, The Netherlands
| | | | - Marcus C de Jong
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Pim de Graaf
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Irshad K, Huang YK, Rodriguez P, Lo J, Aghoghovwia BE, Pan Y, Chang KC. The Neuroimmune Regulation and Potential Therapeutic Strategies of Optic Pathway Glioma. Brain Sci 2023; 13:1424. [PMID: 37891793 PMCID: PMC10605541 DOI: 10.3390/brainsci13101424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/28/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Optic pathway glioma (OPG) is one of the causes of pediatric visual impairment. Unfortunately, there is as yet no cure for such a disease. Understanding the underlying mechanisms and the potential therapeutic strategies may help to delay the progression of OPG and rescue the visual morbidities. Here, we provide an overview of preclinical OPG studies and the regulatory pathways controlling OPG pathophysiology. We next discuss the role of microenvironmental cells (neurons, T cells, and tumor-associated microglia and macrophages) in OPG development. Last, we provide insight into potential therapeutic strategies for treating OPG and promoting axon regeneration.
Collapse
Affiliation(s)
- Khushboo Irshad
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (K.I.); (B.E.A.)
| | - Yu-Kai Huang
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan;
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Paul Rodriguez
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| | - Jung Lo
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
| | - Benjamin E. Aghoghovwia
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (K.I.); (B.E.A.)
| | - Yuan Pan
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (K.I.); (B.E.A.)
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kun-Che Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
- Department of Neurobiology, Center of Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
6
|
Rajagopal R, Khan M, Lethbridge R, Lee G, Lee S, Dyke J, Fabian V, McGrath A, Taylor M, Jacoby P, Endersby R, Nagabushan S, Gottardo NG. Long-term outcomes of symptomatic optic pathway glioma: 32-year experience at a single Western Australian tertiary pediatric oncology center. Front Oncol 2023; 13:1157909. [PMID: 37519788 PMCID: PMC10379632 DOI: 10.3389/fonc.2023.1157909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Optic pathway gliomas (OPGs) are associated with significant risk of visual and endocrine morbidity, but data on long-term outcomes in symptomatic patients is sparse. This study reviews the clinical course, disease progression, survival outcomes and long-term sequelae in pediatric patients with symptomatic OPGs in our institution over three decades. Methods Retrospective review of patients with symptomatic OPG treated in a single tertiary pediatric oncology center from 1984 to 2016. Results A total of 37 patients were diagnosed with symptomatic OPG. Decreased visual acuity was the commonest presenting symptom (75.7%). Surgical intervention was performed in 62.2%; 56.5% underwent biopsy, 26.1% surgical debulking and 17.4% had orbital decompression with cystic fenestration and cosmetic optic nerve excision at different treatment intervals. CSF diversion was performed in 47.8% patients. Histopathologic examination confirmed 86% to be pilocytic astrocytoma and 1 ganglioglioma. 46% received chemotherapy and 48% had radiotherapy, at different intervals. Median follow-up was 13.74 years. In NF1 patients, overall survival (OS) was 100% at 5 years and 55.6 ± 24.8% at 25 years while progression-free-survival (PFS) was 50 ± 15.8% at 5 and 20 years. In non-NF1 patients, OS was 96.2 ± 3.8% at 5 years and 87.4 ± 9% at 25-years. 5-year PFS was 53.8 ± 9.8% and 25-year PFS was 49.0 ± 10%. Cumulative PFS was 53 ± 8.3% at 5 years and 49.7 ± 8.4% at 20 years while cumulative OS was 97.2 ± 2.7% at 5 years and 77.5 ± 10.8% at 25 years. 59.5% patients developed post-operative endocrinopathy. Long-term vision was normal in 8.1%, improved in 13.5%, stabilized in 40.5% but worsened in 37.8% patients. Three patients treated with radiotherapy developed second brain tumors. Conclusion 25-year OS in this cohort was 77.5% but survivorship carried significant long-term morbidities including radiation-induced second malignant brain tumors.
Collapse
Affiliation(s)
- Revathi Rajagopal
- Department of Global Pediatric Medicine, St Jude Children’s Research Hospital, Memphis, TN, United States
| | - Mumtaz Khan
- Department of Anesthesia, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Robert Lethbridge
- School of Pediatrics and Child Health, University of Western Australia, Perth, WA, Australia
| | - Gabriel Lee
- Department of Neurosurgery, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Sharon Lee
- Department of Neurosurgery, Perth Children’s Hospital, Perth, WA, Australia
| | - Jason Dyke
- Department of Neuropathology, Royal Perth Hospital, Perth, WA, Australia
| | - Vicki Fabian
- Department of Neuropathology, Royal Perth Hospital, Perth, WA, Australia
| | - Alycea McGrath
- Department of Radiation Oncology, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Mandy Taylor
- Department of Radiation Oncology, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Peter Jacoby
- Department of Biostatistics, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Raelene Endersby
- Brain Tumor Research Program, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Sumanth Nagabushan
- Kids Cancer Centre, Sydney Children’s Hospital, Sydney, NSW, Australia
- School of Women’s and Children’s Health, University of New South Wales, Sydney, NSW, Australia
| | - Nicholas G. Gottardo
- Brain Tumor Research Program, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
- Department of Pediatric and Adolescent Oncology and Hematology, Perth Children’s Hospital, Perth, WA, Australia
| |
Collapse
|
7
|
Bennebroek CAM, van Zwol J, Porro GL, Oostenbrink R, Dittrich ATM, Groot ALW, Pott JW, Janssen EJM, Bauer NJ, van Genderen MM, Saeed P, Lequin MH, de Graaf P, Schouten-van Meeteren AYN. Impact of Bevacizumab on Visual Function, Tumor Size, and Toxicity in Pediatric Progressive Optic Pathway Glioma: A Retrospective Nationwide Multicentre Study. Cancers (Basel) 2022; 14:cancers14246087. [PMID: 36551572 PMCID: PMC9776082 DOI: 10.3390/cancers14246087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUNDS Bevacizumab (BVZ) is used as a subsequent line of treatment for pediatric optic pathway glioma (OPG) in the case of progression. Data on the treatment effect concerning tumor progression and visual function are scarce and nationwide studies are lacking. METHODS We performed a retrospective, nationwide, multicentre cohort study including all pediatric patients with OPG treated with BVZ in the Netherlands (2009-2021). Progression-free survival, change in visual acuity and visual field, MRI-based radiologic response, and toxicity were evaluated. RESULTS In total, 33 pediatric patients with OPG were treated with BVZ (median 12 months). Visual acuity improved in 20.5%, remained stable in 74.4%, and decreased in 5.1% of 39 of all analysed eyes. The monocular visual field improved in 73.1%, remained stable in 15.4%, and decreased in 7.7% of 25 analysed eyes. Radiologic response at the end of therapy showed a partial response in 7 patients (21.9%), minor response in 7 (21.9%), stable disease in 15 (46.9%), and progressive disease in 3 (9.3%). Progression-free survival at 18 and 36 months after the start of BVZ reduced from 70.9% to 38.0%. Toxicity (≥grade 3 CTCAE) during treatment was observed in five patients (15.2%). CONCLUSION Treatment of BVZ in pediatric patients with OPG revealed stabilisation in the majority of patients, but was followed by progression at a later time point in more than 60% of patients. This profile seems relatively acceptable given the benefits of visual field improvement in more than 70% of analysed eyes and visual acuity improvement in more than 20% of eyes at the cessation of BVZ.
Collapse
Affiliation(s)
- Carlien A. M. Bennebroek
- Department of Ophthalmology, Amsterdam UMC Location University of Amsterdam, 1053 VE Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, 1081 HV Amsterdam, The Netherlands
- Correspondence: ; Tel.: +31-20-5669111
| | - Judith van Zwol
- Department of Ophthalmology, Amsterdam UMC Location University of Amsterdam, 1053 VE Amsterdam, The Netherlands
| | - Giorgio L. Porro
- Department of Ophthalmology Utrecht UMC, 3584 CX Utrecht, The Netherlands
| | - Rianne Oostenbrink
- ENCORE-NF1 Center, Department of General Pediatrics, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Anne T. M. Dittrich
- Department of Pediatrics, Radboud University Medical Center, Amalia Children’s Hospital, 6525 GA Nijmegen, The Netherlands
| | - Annabel L. W. Groot
- Department of Ophthalmology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Jan W. Pott
- Department of Ophthalmology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Etienne J. M. Janssen
- Department of Neurology, Maastricht University Medical Center, 6221 CZ Maastricht, The Netherlands
| | - Noël J. Bauer
- Department of Ophthalmology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
| | - Maria M. van Genderen
- Department of Ophthalmology Utrecht UMC, 3584 CX Utrecht, The Netherlands
- Diagnostic Center for Complex Visual Disorders, Bartiméus, 3703 AJ Zeist, The Netherlands
| | - Peerooz Saeed
- Department of Ophthalmology, Amsterdam UMC Location University of Amsterdam, 1053 VE Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, 1081 HV Amsterdam, The Netherlands
| | - Maarten H. Lequin
- Department of Radiology, Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Pim de Graaf
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, 1081 HV Amsterdam, The Netherlands
| | | |
Collapse
|
8
|
Management of Optic Pathway Glioma: A Systematic Review and Meta-Analysis. Cancers (Basel) 2022; 14:cancers14194781. [PMID: 36230704 PMCID: PMC9563939 DOI: 10.3390/cancers14194781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/25/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Background: OPG accounts for 3−5% of childhood central nervous system (CNS) tumors and about 2% of pediatric glial lesions. Methods: Article selection was performed by searching PubMed, Web of Science, and Cochrane databases. Results: The pooled mortality rate was 0.12 (95%CI 0.09−0.14). Due to the unrepresentative data, improved and not changed outcomes were classified as favorable outcomes and worsened as unfavorable. Meta-analyses were performed to determine the rate of clinical and radiological favorable outcomes. In terms of visual assessment, the pooled rate of a favorable outcome in chemotherapy, radiotherapy, and surgery was 0.74, 0.81, and 0.65, respectively, and the overall pooled rate of the favorable outcome was 0.75 (95%CI 0.70−0.80). In terms of radiological assessment, the rate of a favorable outcome following chemotherapy, radiotherapy, and surgery was 0.71, 0.74, and 0.67, respectively, and the overall pooled rate of the favorable outcome is 0.71 (95%CI 0.65−0.77). The subgroup analysis revealed no significant difference in the rate of clinical and radiological favorable outcomes between the different treatment modalities (p > 0.05). Conclusion: Our analyses showed that each therapeutic modality represents viable treatment options to achieve remission for these patients.
Collapse
|
9
|
Samples DC, Mulcahy Levy JM, Hankinson TC. Neurosurgery for Optic Pathway Glioma: Optimizing Multidisciplinary Management. Front Surg 2022; 9:884250. [PMID: 35599811 PMCID: PMC9114802 DOI: 10.3389/fsurg.2022.884250] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
Optic pathway glioma (OPG) comprises 10% of pediatric brain tumors and 40% of all pediatric low-grade gliomas (pLGGs). While generally considered benign pathologically, many require interventions with chemotherapy, radiation, or targeted therapies. Management has historically foregone tissue diagnosis given the classical clinical/radiographic presentation of these tumors, inability to safely remove the lesions surgically, and efficacy and safety of available chemotherapy options. Furthermore, when considering such aspects as their delicate location, the role of surgery continues to be heavily debated. More recently, however, a greater understanding of the genetic drivers of OPGs has made operative tissue sampling a critical step in management planning, specifically for patients without Neurofibromatosis, Type I (NF1). Given the need for long-term, complex management of pediatric OPGs, it is crucial that a multidisciplinary approach is employed, and the rapidly expanding role of molecular characterization be incorporated into their management.
Collapse
Affiliation(s)
- Derek C. Samples
- Department of Neurosurgery, Children’s Hospital Colorado, Aurora, CO, United States
- Correspondence: Derek C. Samples
| | - Jean M. Mulcahy Levy
- Department of Pediatrics (Center for Cancer and Blood Disorders), University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, United States
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, United States
| | - Todd C. Hankinson
- Department of Neurosurgery, Children’s Hospital Colorado, Aurora, CO, United States
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, United States
| |
Collapse
|
10
|
Bennebroek CAM, Wijninga LE, Limpens J, Schouten-van Meeteren AYN, Saeed P. Impact of systemic anticancer therapy in pediatric optic pathway glioma on visual function: A systematic review. PLoS One 2021; 16:e0258548. [PMID: 34673789 PMCID: PMC8530362 DOI: 10.1371/journal.pone.0258548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 09/29/2021] [Indexed: 01/11/2023] Open
Abstract
Pediatric optic pathway glioma (OPG) can seriously decrease visual function in the case of progression. Systemic anticancer therapy (SAT) is considered the treatment of first choice for unresectable OPG. New SAT modalities for the treatment of progressive OPG have been introduced in the last decade, including VEGF and MAPK pathway inhibition. This systematic review evaluated the effect of SAT on change in visual acuity and visual field in OPG. A systematic review was performed on SAT for OPG (January 1990 to August 2020). MEDLINE and EMBASE (Ovid) were searched for studies reporting on change in visual acuity and visual field after treatment with SAT for OPG. Overall, 11 series, including 358 patients, fulfilled the eligibility criteria. After follow-up of median 3.7 years (range: cessation of SAT- 8.2 years), improvement in binocular VA was found in 0-45% of studies, stability in 18-77% and a decrease in 0-82%. Two studies reported on change in visual field (improvement in 19% and 71% of patients), although either the change was not defined or the testing strategy was lacking. Considerable heterogeneity was present among the included studies, such as variety in the combinations of SAT administered, status of neurofibromatosis type 1, definition regarding change in visual acuity, 1- or 2-eye analysis, diversity in anatomic location, and extent of follow-up, all of which made meta-analysis inappropriate. This systematic review suggests that the impact of SAT in OPG on visual function is still unclear. The wide ranges reported on the efficacy of SAT and the observed heterogeneity highlight the need for prospective studies with uniform definitions of outcome parameters.
Collapse
Affiliation(s)
- Carlien A. M. Bennebroek
- Department of Ophthalmology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Laura. E. Wijninga
- Department of Ophthalmology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jaqueline Limpens
- Medical Library, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Peerooz Saeed
- Department of Ophthalmology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Thirunavu VM, Mohammad LM, Kandula V, Beestrum M, Lam SK. Vision Outcomes for Pediatric Patients With Optic Pathway Gliomas Associated With Neurofibromatosis Type I: A Systematic Review of the Clinical Evidence. J Pediatr Hematol Oncol 2021; 43:135-143. [PMID: 33480655 DOI: 10.1097/mph.0000000000002060] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/08/2020] [Indexed: 01/14/2023]
Abstract
Children with neurofibromatosis type I (NF1) have a higher predisposition for low-grade astrocytomas of the optic pathway, commonly referred to as optic pathway gliomas (OPGs). OPGs can result in visual deterioration. Treatment outcomes in OPG-NF1 management are often reported around tumor stabilization. We sought to compare vision outcomes associated with different OPG treatment strategies to inform about this important functional metric. A meta-analysis exploring the different modalities to treat children with OPG-NF1 was conducted following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines using multiple databases. Of the 113 articles identified in the search, 23 full text articles, representing 564 patients, were included for review. These articles included retrospective, prospective, and randomized controlled studies on observation (n=9), chemotherapy (n=19), radiation therapy (n=6), and surgery (n=7). Of the patients undergoing observation, 87% (60/69) demonstrated stable acuity. In the chemotherapy studies, 27.3% (72/264) demonstrated improved acuity/visual field and/or visual-evoked potential amplitudes, 39.4% (104/264) stable acuity, and 33.3% (88/264) deterioration. Both the radiation and surgical treatments reported worsening acuity at 90.9% (10/11) and 73.3% (11/15), respectively. Causal associations are not known. Indications for and timing of treatment choice warrant larger scale study to provide further understanding.
Collapse
Affiliation(s)
| | - Laila M Mohammad
- Department of Neurological Surgery, Division of Pediatric Neurosurgery
| | - Viswajit Kandula
- Department of Neurological Surgery, Division of Pediatric Neurosurgery
| | - Molly Beestrum
- Galter Health Sciences Library, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Sandi K Lam
- Department of Neurological Surgery, Division of Pediatric Neurosurgery
| |
Collapse
|
12
|
Hanania AN, Paulino AC, Ludmir EB, Shah VS, Su JM, McGovern SL, Baxter PA, McAleer MF, Grosshans DR, Okcu MF, Chintagumpala MM. Early radiotherapy preserves vision in sporadic optic pathway glioma. Cancer 2021; 127:2358-2367. [PMID: 33739455 DOI: 10.1002/cncr.33497] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/02/2021] [Accepted: 01/26/2021] [Indexed: 01/15/2023]
Abstract
BACKGROUND Sporadic optic pathway/hypothalamic gliomas represent a unique entity within pediatric low-grade glioma. Despite favorable survival, location makes treatment difficult and local progression debilitating. This study is a longitudinal assessment of visual acuity (VA) among children treated within the last 2 decades. METHODS Clinical characteristics were abstracted for patients treated from 2000 to 2018 at Texas Children's Cancer Center in Houston. Ophthalmologic data taken at 3- to 6-month intervals were examined with age-appropriate VA metrics converted to the LogMAR (logarithm of the minimum angle of resolution) scale. Kaplan-Meier blindness-free survival (BFS) curves, calculated as time-to-bilateral functional blindness (LogMAR ≥0.8 in both eyes), were calculated for patients receiving early radiation therapy (RT; upfront or as first-line salvage treatment) or chemotherapy (CT) and evaluated using the log-rank test. RESULTS Thirty-eight patients with a median follow-up of 8.5 years (range, 2-17 years) were identified. Median age at diagnosis was 3 years (interquartile range, <1-6 years). Early RT was administered in 11 patients (29%). Twenty-seven patients (71%) were treated primarily with CT, initiated at a median age of 3.5 years (range, <1-11 years). Eight patients in the CT group did eventually require RT secondary to VA loss and following multiple lines of CT. Median age at RT for all patients was 11 years (range, 3-17 years). BFS rates were 81% at 5 years and 60% at 8 years for CT and 100% at 5 and 8 years for early RT (P = .017). CONCLUSIONS In a contemporary cohort, early RT, defined as initial or first-line salvage therapy, was found to have superior BFS for appropriately selected patients with sporadic optic pathway/hypothalamic gliomas. LAY SUMMARY Children with low-grade brain tumors of the optic pathway generally have excellent long-term survival; however, given the location of these tumors, there can commonly be threatened vision if the tumor grows. Although radiation is generally deferred in children on the basis of legitimate concerns regarding the effects on the developing brain, it may represent a vision-preserving therapy for well-selected older patients.
Collapse
Affiliation(s)
- Alexander N Hanania
- Department of Radiation Oncology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Arnold C Paulino
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ethan B Ludmir
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Veeral S Shah
- Department of Pediatric Ophthalmology, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Jack M Su
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Susan L McGovern
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patricia A Baxter
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Mary Frances McAleer
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David R Grosshans
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - M Fatih Okcu
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas
| | | |
Collapse
|
13
|
MRI-based diagnosis and treatment of pediatric brain tumors: is tissue sample always needed? Childs Nerv Syst 2021; 37:1449-1459. [PMID: 33821340 PMCID: PMC8084800 DOI: 10.1007/s00381-021-05148-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/24/2021] [Indexed: 11/23/2022]
Abstract
Traditional management of newly diagnosed pediatric brain tumors (PBTs) consists of cranial imaging, typically magnetic resonance imaging (MRI), and is frequently followed by tissue diagnosis, through either surgical biopsy or tumor resection. Therapy regimes are typically dependent on histological diagnosis. To date, many treatment regimens are based on molecular biology. The scope of this article is to discuss the role of diagnosis and further treatment of PBTs based solely on MRI features, in light of the latest treatment protocols. Typical MRI findings and indications for surgical biopsy of these lesions are described.
Collapse
|
14
|
Attina G, Maurizi P, Triarico S, Capozza MA, Romano A, Mastrangelo S, Ruggiero A. Management of Children with Optic Gliomas and Neurofibromatosis Type 1. BIOMEDICAL AND PHARMACOLOGY JOURNAL 2020; 13:1601-1606. [DOI: 10.13005/bpj/2035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Optic pathway gliomas (OPG) are a common cancer in children with neurofibromatosis type 1. OPGs can cause clinical symptoms such as reduction of visual acuity, alterations of the visual field, pallor of the optical papilla, strabismus, endocrinological alterations up to diencephalic syndrome.The current guidelines provide for wait and see as the main approach if the tumor is not causing visual deterioration and adopting treatment only in the event of significant impairment of the visual function. Therefore, it is essential to early detect the visual deterioration changes as well as the identification of children eligible for treatment.
Collapse
Affiliation(s)
- Giorgio Attina
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Palma Maurizi
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Silvia Triarico
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Michele Antonio Capozza
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Alberto Romano
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Stefano Mastrangelo
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Antonio Ruggiero
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| |
Collapse
|
15
|
Stella I, Helleringer M, Joud A, Chastagner P, Thomas R, Klein O. Optic pathway tumor in children: Toward a new classification for neurosurgical use. Neurochirurgie 2020; 67:336-345. [PMID: 33232713 DOI: 10.1016/j.neuchi.2020.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 10/26/2020] [Accepted: 11/11/2020] [Indexed: 11/26/2022]
Abstract
OBJECT Optic pathway tumors (OPT) represent a challenge for pediatric neurosurgeons. Role of surgery is debated due to the high risk of iatrogenic damage, and in lasts decades it lost its importance in favor of chemotherapy. However, in some cases surgery is necessary to make biomolecular and histological diagnosis, to manage intracranial hypertension (IH) and to cooperate with medical therapies in controlling tumor relapse. With the aim to standardize selection of surgical OPT cases, we propose a simple, practical and reproducible classification. METHODS We retrospectively analyzed data of 38 patients with OPT treated at our institution (1990-2018). After careful analysis of MRI images, we describe a new classification system. Group 1: lesion limited to one or both optic nerve(s). Group 2: chiasmatic lesions extending minimally to hypothalamus. Group 3: hypothalamo-chiasmatic exophitic lesions invading the third ventricle; they can be further divided on the base of concomitant hydrocephalus. Group 4: hypothalamo-chiasmatic lesions extending widely in lateral direction, toward the temporal or the frontal lobes. Patients' data and adopted treatment are reported and analyzed, also depending on this classification. RESULTS Twenty children were operated on for treatment of OPT during the study period. Permanent clinical impairment was noted in 5 (25%) of operated patients, while visual improvement was noted in 1 patient. OS rate was 100% at 5 years, with a median follow up of 9 years (ranging from 2 to 23). Prevalence of intracranial hypertension and proportion of first-line surgical treatment decision were significantly higher in groups 3-4 compared to groups 1-2 (P<0.001 for both tests). CONCLUSION Surgery can offer a valuable therapeutic complement for OPT without major risk of iatrogenic damage. Surgery is indispensable in cases presenting with IH, as in groups 3 and 4 lesions. Eligibility of patients to surgery can be based on this new classification system.
Collapse
Affiliation(s)
- I Stella
- Pediatric Neurosurgical Unit, Nancy Regional University Hospital, University of Lorraine, France.
| | - M Helleringer
- Pediatric Neurosurgical Unit, Nancy Regional University Hospital, University of Lorraine, France
| | - A Joud
- Pediatric Neurosurgical Unit, Nancy Regional University Hospital, University of Lorraine, France
| | - P Chastagner
- Pediatric Onco-hematology Department, Nancy Regional University Hospital, University of Lorraine, France
| | - R Thomas
- Methodologic, data-management and statistics Unit, Nancy Regional University Hospital, University of Lorraine, France
| | - O Klein
- Pediatric Neurosurgical Unit, Nancy Regional University Hospital, University of Lorraine, France
| |
Collapse
|
16
|
Visual function tests including the role of optical coherence tomography in neurofibromatosis 1. Childs Nerv Syst 2020; 36:2363-2375. [PMID: 32749524 DOI: 10.1007/s00381-020-04706-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/25/2020] [Indexed: 10/23/2022]
Abstract
Optic pathway glioma (OPG) is a common and significant complication of neurofibromatosis 1 (NF-1) that might lead to vision loss. The main reason to treat OPG is to preserve vision. Tumor location along the visual pathway largely dictates the presenting signs and symptoms. Clinical ophthalmic evaluation is focused on optic nerve functions including evaluation of pupils' reaction to light, visual acuity, color vision, and visual field, as well as optic nerve appearance. An important relatively new ancillary test is optic coherence tomography (OCT) that measures the volume of retinal nerve fiber layer around the optic nerve and the ganglion cell layer-inner plexiform layer (GCL-IPL) of the macula, both proved to be strongly associated with losing vision in OPG. Accurate evaluation of vision functions plays a critical role in the decision of treatment. In this review, we describe the ophthalmological assessment including new biomarkers in clinical use. We also outline prognostic factors and current recommendations for surveillance and indications for treatment.
Collapse
|
17
|
Shofty B, Ben Sira L, Constantini S. Neurofibromatosis 1-associated optic pathway gliomas. Childs Nerv Syst 2020; 36:2351-2361. [PMID: 32524182 DOI: 10.1007/s00381-020-04697-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 05/21/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Optic Pathway Gliomas (OPG) are the most common brain tumor in Neurofibromatosis 1 patients (NF1). They are found along the optic pathway and may involve the optic nerves, chiasm, retro-chiasmatic structures, and the optic radiations. NF1 associate OPG (NF1-OPG) have variable presentation, disease course and response to treatment. The optimal management is patient-specific and should be tailored by a multidisciplinary team. Age, sex, histology, and molecular markers may be important factors in the individualized decision-making process. Chemotherapy is the first-line treatment in cases of progressive tumors, and visual preservation is the main goal of treatment. PURPOSE In this paper we will review the disease, practical management, and recent advances of NF1-OPG.
Collapse
Affiliation(s)
- Ben Shofty
- Department of Neurosurgery, Tel-Aviv Medical Center, The Gilbert Israeli International Neurofibromatosis Center (GIINFC), Tel Aviv University, Tel Aviv, Israel
| | - Liat Ben Sira
- Pediatric Radiology, Tel-Aviv Medical Center, The Gilbert Israeli International Neurofibromatosis Center (GIINFC), Tel Aviv University, Tel Aviv, Israel
| | - Shlomi Constantini
- Department of Pediatric Neurosurgery, Dana Children's Hospital, Tel-Aviv Medical Center, The Gilbert Israeli International Neurofibromatosis Center (GIINFC), Tel Aviv University, 6th Weizmann St., 64239, Tel-Aviv, Israel.
| |
Collapse
|
18
|
Bates JE, Indelicato DJ, Morris CG, Rotondo RL, Bradley JA. Visual decline in pediatric survivors of brain tumors following radiotherapy. Acta Oncol 2020; 59:1257-1262. [PMID: 32762400 DOI: 10.1080/0284186x.2020.1803500] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE Radiotherapy-related visual decline is a significant concern in survivors of childhood cancer; however, data establishing the dose-response relationship between dose to the optic apparatus and visual acuity decline in children are sparse. We aimed to determine this relationship in a cohort of children treated with proton therapy. MATERIAL AND METHODS We identified 458 children with 875 eyes at risk treated with proton therapy for intracranial malignancy between December 2006 and September 2018. Eyes were considered at risk if either the ipsilateral optic nerve or optic chiasm received ≥30 GyRBE to 0.1 cm3. Kaplan-Meier and Normal Tissue Complication Probability modeling was used to establish the relationship between radiotherapy dose and risk of visual decline. RESULTS Excluding children with tumor progression, no patient experienced complete vision loss. The actuarial 5-year rate of any visual acuity decline was 2.6% (95% confidence interval [CI]: 1.5%-4.6%). The dose to 0.1 cm3 of the ipsilateral optic nerve or optic chiasm resulting in a 1%, 5%, and 10% risk of acuity decline were 52.7 GyRBE, 56.6 GyRBE, and 58.3 GyRBE. Visual decline was only seen in children with primary tumors of the optic pathway or suprasellar region. CONCLUSIONS Visual acuity decline following radiotherapy for intracranial malignancies in children is rare. A dose of approximately 56 GyRBE to 0.1 cm3 results in an approximately 5% risk of visual acuity decline for children with suprasellar or optic pathway tumors. A dose to 0.1 cm3 of 56 GyRBE appears to be safe for children with tumors elsewhere in the brain.
Collapse
Affiliation(s)
- James E. Bates
- Department of Radiation Oncology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Daniel J. Indelicato
- Department of Radiation Oncology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Christopher G. Morris
- Department of Radiation Oncology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Ronny L. Rotondo
- Department of Radiation Oncology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Julie A. Bradley
- Department of Radiation Oncology, University of Florida College of Medicine, Jacksonville, FL, USA
| |
Collapse
|
19
|
Liao C, Zhang H, Liu Z, Han Z, Li C, Gong J, Liu W, Ma Z, Tian Y. The Visual Acuity Outcome and Relevant Factors Affecting Visual Improvement in Pediatric Sporadic Chiasmatic-Hypothalamic Glioma Patients Who Received Surgery. Front Neurol 2020; 11:766. [PMID: 32973646 PMCID: PMC7466562 DOI: 10.3389/fneur.2020.00766] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/22/2020] [Indexed: 11/29/2022] Open
Abstract
Background: The role and effectiveness of primary surgical treatment for sporadic chiasmatic–hypothalamic glioma (CHG) are not clear. The present study was to describe sporadic CHG visual acuity (VA) outcomes after surgery and to analyze the relevant factors affecting VA improvement. Methods: Forty-five pediatric sporadic CHG patients who met the inclusion criteria were included in a retrospective study. All patients received primary intratumor partial resection. Disease characteristics, treatment strategies, complications, and VA outcome were analyzed. Univariate and multivariate analyses were performed to identify relevant factors of VA improvement. Receiver operating characteristic (ROC) analysis was performed to evaluate the predictive accuracy of measurement indexes. Results: There were 77 eyes of 45 children suffering from various levels of VA impairment before surgical treatment, and only 13 eyes had normal vision. Patients with resection extents >70, 50–70, and <50% accounted for 26.67, 24.44, and 48.89%, respectively. The percentages of VA maintained and deteriorated in normal vision eyes were 61.54 and 38.46%. The percentages of VA improved, maintained, and deteriorated in visually impaired eyes after surgery were 29.87, 45.45, and 24.68%, respectively. There was a positive correlation between the IVA level and VA improvement. There was no significant difference in VA improvement between the different resection extents. Blindness occurred in ~4.4%. Approximately 11.1% of the children had complications that affected quality of life, which correlated with resection extent. IVA and tumor size were correlated with VA improvement. The AUC for IVA + tumor size predicting VA improvement was 0.831. The cutoff points for IVA level and tumor volume were 4.5 and 43.50 cm3, respectively. Conclusions: IVA and tumor size were correlated with VA improvement after primary intratumor partial resection. Children with IVA ≥ level 5 were more likely to achieve visual improvement after decompression surgery, but decompression had limited effectiveness on vision improvement in patients with tumor volumes ≥ 43.50 cm3. Performing resections < 50% was safe and did not reduce the effect of decompression to improve VA.
Collapse
Affiliation(s)
- Chihyi Liao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Brain Tumor, China National Clinical Research Center for Neurological Diseases, Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Heng Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Brain Tumor, China National Clinical Research Center for Neurological Diseases, Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Zhiming Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Brain Tumor, China National Clinical Research Center for Neurological Diseases, Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Zhe Han
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Brain Tumor, China National Clinical Research Center for Neurological Diseases, Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Chunde Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Brain Tumor, China National Clinical Research Center for Neurological Diseases, Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Jian Gong
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Brain Tumor, China National Clinical Research Center for Neurological Diseases, Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Wei Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Brain Tumor, China National Clinical Research Center for Neurological Diseases, Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Zhenyu Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Brain Tumor, China National Clinical Research Center for Neurological Diseases, Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Yongji Tian
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Brain Tumor, China National Clinical Research Center for Neurological Diseases, Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
20
|
Zahavi A, Luckman J, Ben-David GS, Toledano H, Michowiz S, Vardizer Y, Goldenberg-Cohen N. Proptosis due to intraorbital space-occupying lesions in children. Graefes Arch Clin Exp Ophthalmol 2020; 258:2541-2550. [PMID: 32676791 DOI: 10.1007/s00417-020-04840-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 07/05/2020] [Accepted: 07/10/2020] [Indexed: 10/23/2022] Open
Abstract
PURPOSE To report the 10-year experience of two tertiary medical centers with children presenting with proptosis due to an intraorbital space-occupying lesion. METHODS Patients were identified by file review. Data were collected on demographics, findings on ophthalmologic and imaging evaluations, etiology, treatment, and outcome. RESULTS Nineteen children (7 male) were included. Eleven patients had optic nerve glioma, including 9 with substantially decreased visual acuity. Treatment consisted of chemotherapy alone or with radiation, resection or anti-VEGF agents, MEK inhibitor, or observation only (n = 1). Visual and cosmetic outcomes were poor in all cases. Outcome for arteriovenous malformations was good following corticosteroid treatment (n = 1), but catheterization led to persistent proptosis and fluctuating visual acuity (n = 1). Compound capillary hemangioma (n = 1) was treated with laser and systemic beta blockers with satisfactory results. Rhabdomyosarcoma had a good prognosis in one patient treated with resection and radiation but was fatal in another even after chemotherapy. Juvenile xanthogranuloma, frontal bone osteoma, and localized hypertrophic neuropathy of the supraorbital nerve (n = 1 each) were treated by resection with good visual and cosmetic outcomes. CONCLUSIONS Proptosis accompanied by visual loss is an uncommon presentation in children and suggests an orbital tumor. We found that visual outcome was better when the nerve was not involved by tumor. Optic nerve glioma was the most common cause and failed to respond to various treatments. Catheterization for arteriovenous malformation did not prevent proptosis, and final visual acuity fluctuated. Surgery for rhabdomyosarcoma and xanthogranuloma led to remission with preservation of vision in 2 of 3 cases.
Collapse
Affiliation(s)
- Alon Zahavi
- Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel.,Department of Ophthalmology, Rabin Medical Center, Beilinson Hospital, 4941492, Petah Tikva, Israel.,Krieger Eye Research Laboratory, Felsenstein Medical Research Center, 4941492, Petah Tikva, Israel
| | - Judith Luckman
- Department of Radiology, Rabin Medical Center, Beilinson Hospital, 4941492, Petah Tikva, Israel
| | - Geulah S Ben-David
- Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Helen Toledano
- Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel.,Department of Oncology, Schneider Children's Medical Center of Israel, 49420235, Petah Tikva, Israel
| | - Shalom Michowiz
- Department of Neurosurgery, Hadassah Medical Center, 91120, Jerusalem, Israel
| | - Yoav Vardizer
- Department of Ophthalmology, Bnai-Zion Medical Center, 3339419, Haifa, Israel
| | - Nitza Goldenberg-Cohen
- Krieger Eye Research Laboratory, Felsenstein Medical Research Center, 4941492, Petah Tikva, Israel. .,Department of Ophthalmology, Bnai-Zion Medical Center, 3339419, Haifa, Israel. .,Technion - Israel Institute of Technology, 3200003, Haifa, Israel.
| |
Collapse
|
21
|
Abstract
Phakomatoses present with characteristic findings on the skin, central or peripheral nervous system, and tumors. Neurofibromatosis type 1 is the most common syndrome and is characterized by Café-au-lait macules, intertriginous freckling, Lisch nodules, and tumors including neurofibromas, malignant peripheral nerve sheath tumors, and gliomas. Tuberous Sclerosis Complex is characterized by benign hamartomas presenting with hypomelanotic macules, shagreen patches, angiofibromas, confetti lesions and tumors including cortical tubers, subependymal nodules, subependymal giant cell astrocytomas and tumors of the kidney, lung, and heart. Managing these disorders requires disease specific supportive care, tumor monitoring, surveillance for selected cancers, and treatment of comorbid conditions.
Collapse
Affiliation(s)
- Benjamin Becker
- Department of Neurology, Wake Forest Baptist Health, 1 Medical Center Boulevard, Winston Salem, NC 27157, USA.
| | - Roy E Strowd
- Department of Neurology, Wake Forest Baptist Health, 1 Medical Center Boulevard, Winston Salem, NC 27157, USA; Department of Internal Medicine, Section on Hematology and Oncology, Wake Forest Baptist Health, Winston Salem, NC 27157, USA; Translational Science Institute, Wake Forest Baptist Health, Winston Salem, NC 27157, USA
| |
Collapse
|
22
|
Optic Pathway Glioma in Type 1 Neurofibromatosis: Review of Its Pathogenesis, Diagnostic Assessment, and Treatment Recommendations. Cancers (Basel) 2019; 11:cancers11111790. [PMID: 31739524 PMCID: PMC6896195 DOI: 10.3390/cancers11111790] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/12/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022] Open
Abstract
Type 1 neurofibromatosis (NF1) is a dominantly inherited condition predisposing to tumor development. Optic pathway glioma (OPG) is the most frequent central nervous system tumor in children with NF1, affecting approximately 15-20% of patients. The lack of well-established prognostic markers and the wide clinical variability with respect to tumor progression and visual outcome make the clinical management of these tumors challenging, with significant differences among distinct centers. We reviewed published articles on OPG diagnostic protocol, follow-up and treatment in NF1. Cohorts of NF1 children with OPG reported in the literature and patients prospectively collected in our center were analyzed with regard to clinical data, tumor anatomical site, diagnostic workflow, treatment and outcome. In addition, we discussed the recent findings on the pathophysiology of OPG development in NF1. This review provides a comprehensive overview about the clinical management of NF1-associated OPG, focusing on the most recent advances from preclinical studies with genetically engineered models and the ongoing clinical trials.
Collapse
|
23
|
van Baarsen K, Roth J, Serova N, Packer RJ, Shofty B, Thomale UW, Cinalli G, Toledano H, Michowiz S, Constantini S. Optic pathway-hypothalamic glioma hemorrhage: a series of 9 patients and review of the literature. J Neurosurg 2019; 129:1407-1415. [PMID: 29424646 DOI: 10.3171/2017.8.jns163085] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/08/2017] [Indexed: 11/06/2022]
Abstract
OBJECTIVEHemorrhage (also known as apoplexy) in optic pathway gliomas (OPGs) is rare. Because of the variable presentations and low incidence of OPG hemorrhages, little is known about their clinical course and the best treatment options. The aim of this work was to review risk factors, clinical course, and treatment strategies of optic glioma hemorrhages in the largest possible number of cases.METHODSA total of 34 patients were analyzed. Nine new cases were collected, and 25 were identified in the literature. Data regarding demographics, radiological and histological features, treatment, and outcome were retrospectively reviewed.RESULTSThe majority of patients were younger than 20 years. Only 3 patients were known to have neurofibromatosis. The histopathological diagnosis was pilocytic astrocytoma in the majority of cases. Five patients had intraorbital hemorrhages, whereas 29 patients had intracranial hemorrhage; the majority of intracranial bleeds were treated surgically. Six patients, all with intracranial hemorrhage, died due to recurrent bleeding, hydrocephalus, or surgical complications. No clear risk factors could be identified.CONCLUSIONSIntracerebral OPG hemorrhages have a fatal outcome in 20% of cases. Age, hormonal status, neurofibromatosis involvement, and histopathological diagnosis have been suggested as risk factors for hemorrhage, but this cannot be reliably established from the present series. The goals of surgery should be patient survival and prevention of further neurological and ophthalmological deterioration.
Collapse
Affiliation(s)
- Kirsten van Baarsen
- 1Department of Pediatric Neurosurgery, International Israel Neurofibromatosis Center (IINFC), Dana Children's Hospital, Tel Aviv Medical Center, Tel Aviv University, Israel.,2Department of Neurosurgery, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Jonathan Roth
- 1Department of Pediatric Neurosurgery, International Israel Neurofibromatosis Center (IINFC), Dana Children's Hospital, Tel Aviv Medical Center, Tel Aviv University, Israel
| | - Natalia Serova
- 3Department of Neuro-ophthalmology, Burdenko Neurosurgical Institute, Moscow, Russia
| | - Roger J Packer
- 4Center for Neuroscience and Behavioral Medicine, Children's National Medical Center, Washington, DC
| | - Ben Shofty
- 1Department of Pediatric Neurosurgery, International Israel Neurofibromatosis Center (IINFC), Dana Children's Hospital, Tel Aviv Medical Center, Tel Aviv University, Israel
| | - Ulrich-W Thomale
- 5Department of Pediatric Neurosurgery, Charité Universitätsmedizin, Berlin, Germany
| | - Giuseppe Cinalli
- 6Department of Neurosurgery, Santobono-Pausilipon Children's Hospital, Naples, Italy; and
| | - Helen Toledano
- Departments of7Pediatric Oncology and.,8Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - Shalom Michowiz
- 8Sackler Faculty of Medicine, Tel Aviv University, Israel.,9Pediatric Neurosurgery, Schneider Children's Medical Center of Israel, Petach Tikva; and
| | - Shlomi Constantini
- 1Department of Pediatric Neurosurgery, International Israel Neurofibromatosis Center (IINFC), Dana Children's Hospital, Tel Aviv Medical Center, Tel Aviv University, Israel
| |
Collapse
|
24
|
Cooney T, Yeo KK, Kline C, Prados M, Haas-Kogan D, Chi S, Mueller S. Neuro-Oncology Practice Clinical Debate: targeted therapy vs conventional chemotherapy in pediatric low-grade glioma. Neurooncol Pract 2019; 7:4-10. [PMID: 32257279 DOI: 10.1093/nop/npz033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The treatment of children with low-grade glioma has evolved over the last several decades, beginning initially with focal radiotherapy, which has now been largely replaced by systemic treatment with conventional chemotherapy agents or more recently molecularly targeted therapeutics. A consensus standard of care is not well defined, leaving clinicians and parents to choose from an increasing number of options, often without complete information concerning the associated risks and benefits. Issues critical to this topic include timing of interventions (when to treat), preservation of neurological function (goals of treatment), choice of initial therapy strategy (conventional cytotoxic chemotherapy vs molecularly targeted therapy), duration of treatment (how long, and what clinical or imaging endpoints to consider), and perhaps most important, risk reduction relative to anticipated benefit. The groups from the University of California, San Francisco and Dana Farber Cancer Institute, moderated by Michael Prados, herein debate the merits of cytotoxic chemotherapy and targeted therapeutics as initial treatment strategies in pediatric low-grade glioma, a topic discussed daily in Tumor Boards across the United States and abroad. Prospective, randomized, phase 3 trials comparing the 2 strategies, conducted within homogenous disease settings, with consistently evaluated functional and imaging endpoints, are not available to guide the risks/benefit discussion. As is often the case in rare biologically diverse diseases, in a vulnerable population, therapy decisions are frequently based on incomplete data, physician experience, bias to some degree, and patient/family preference.
Collapse
Affiliation(s)
- Tabitha Cooney
- Department of Pediatrics, University of California, San Francisco
| | - Kee Kiat Yeo
- Dana Farber Cancer Institute, Boston, MA.,Boston Children's Hospital, MA.,Harvard Medical School, Boston MA
| | - Cassie Kline
- Department of Pediatrics, University of California, San Francisco.,Department of Neurology, University of California, San Francisco
| | - Michael Prados
- Department of Neurology, University of California, San Francisco.,Department of Neurosurgery, University of California, San Francisco
| | - Daphne Haas-Kogan
- Boston Children's Hospital, MA.,Department of Radiation Oncology, Brigham and Women's Hospital, Boston, MA.,Harvard Medical School, Boston MA
| | - Susan Chi
- Dana Farber Cancer Institute, Boston, MA.,Boston Children's Hospital, MA.,Harvard Medical School, Boston MA
| | - Sabine Mueller
- Department of Pediatrics, University of California, San Francisco.,Department of Neurology, University of California, San Francisco.,Department of Neurosurgery, University of California, San Francisco.,Children's Hospital University of Zürich, Oncology, Switzerland
| |
Collapse
|
25
|
|
26
|
Calixto NC, Simão GN, Dos Santos AC, de Oliveira RS, Junior LGD, Valera ET, Cintra MB, Mello AS. Monitoring optic chiasmatic-hypothalamic glioma volumetric changes by MRI in children under clinical surveillance or chemotherapy. Childs Nerv Syst 2019; 35:63-72. [PMID: 30078056 DOI: 10.1007/s00381-018-3904-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 07/04/2018] [Indexed: 10/28/2022]
Abstract
PURPOSE Optic pathway gliomas represent 5% of pediatric brain tumors and are typically low-grade lesions. Because of their unpredictable clinical course, adequate treatment approaches have been controversial, involving surveillance, surgery, chemotherapy, and radiotherapy. In this study, we use volumetric imaging to compare evolution of optic chiasmatic-hypothalamic gliomas (OCHG) treated with and without chemotherapy, analyzing tumor volume variation during the overall period. METHODS A total of 45 brain MRI were retrospectively analyzed for 14 patients with OCHG. Volumetric assessment of the lesions was performed by a neuroradiologist, using software DISPLAY. OCHG patients were allocated into two groups: group 1 (n = 8) who underwent chemotherapy and group 2 (n = 6) who did not receive chemotherapy. Outcome analysis was performed comparing tumor volume evolution of these two groups. RESULTS The results showed a reduction of 4.4% of the volume of the lesions for group 1 after the end of chemotherapy, with an increase of 5.3% in volume in the late follow-up examination. For group 2, we found a slight reduction (5%) of the overall volume of the lesions, both with no statistical significance (p > 0.05). CONCLUSIONS From the limited series analyzed in this study, no significant differences were observed in relation to the volume change of lesions treated or not treated with chemotherapy. Larger prospective clinical trials are needed to better evaluate the effect of chemotherapy and radiological response of OCHG.
Collapse
Affiliation(s)
- Nathalia Cunha Calixto
- Division of Radiology, Department of Clinics, University Hospital of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049-900, Brazil.
| | - Gustavo Novelino Simão
- Division of Radiology, Department of Clinics, University Hospital of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049-900, Brazil
| | - Antonio Carlos Dos Santos
- Division of Radiology, Department of Clinics, University Hospital of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049-900, Brazil
| | - Ricardo Santos de Oliveira
- Division of Pediatric Neurosurgery, Department of Surgery and Anatomy, University Hospital of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049-900, Brazil
| | - Luiz Guilherme Darrigo Junior
- Division of Pediatric Neuroncology, Department of Pediatrics, University Hospital of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049-900, Brazil
| | - Elvis Terci Valera
- Division of Pediatric Neuroncology, Department of Pediatrics, University Hospital of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049-900, Brazil
| | - Murilo Bicudo Cintra
- Division of Radiology, Department of Clinics, University Hospital of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049-900, Brazil
| | - Alessandro Spano Mello
- Division of Radiology, Department of Clinics, University Hospital of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049-900, Brazil
| |
Collapse
|
27
|
Kinori M, Hodgson N, Zeid JL. Ophthalmic manifestations in neurofibromatosis type 1. Surv Ophthalmol 2018; 63:518-533. [PMID: 29080631 DOI: 10.1016/j.survophthal.2017.10.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 10/18/2022]
Abstract
Neurofibromatosis type 1 (NF1) is a relatively common multisystemic inherited disease and has been extensively studied by multiple disciplines. Although genetic testing and confirmation are available, NF1 remains a clinical diagnosis. Many manifestations of NF1 involve the eye and orbit, and the ophthalmologist, therefore, plays a significant role in the diagnosis and treatment of NF1 patients. Improvements in diagnostic and imaging instruments have provided new insight to study the ophthalmic manifestations of the disease. We provide a comprehensive and up-to-date overview of the ocular and orbital manifestations of NF1.
Collapse
Affiliation(s)
- Michael Kinori
- Department of Ophthalmology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Nickisa Hodgson
- Department of Ophthalmology, Shiley Eye Institute, University of California, San Diego, California, USA
| | - Janice Lasky Zeid
- Department of Ophthalmology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA.
| |
Collapse
|
28
|
Freret ME, Gutmann DH. Insights into optic pathway glioma vision loss from mouse models of neurofibromatosis type 1. J Neurosci Res 2018; 97:45-56. [PMID: 29704429 DOI: 10.1002/jnr.24250] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 04/09/2018] [Indexed: 12/12/2022]
Abstract
Neurofibromatosis type 1 (NF1) is a common cancer predisposition syndrome caused by mutations in the NF1 gene. The NF1-encoded protein (neurofibromin) is an inhibitor of the oncoprotein RAS and controls cell growth and survival. Individuals with NF1 are prone to developing low-grade tumors of the optic nerves, chiasm, tracts, and radiations, termed optic pathway gliomas (OPGs), which can cause vision loss. A paucity of surgical tumor specimens and of patient-derived xenografts for investigative studies has limited our understanding of human NF1-associated OPG (NF1-OPG). However, mice genetically engineered to harbor Nf1 gene mutations develop optic gliomas that share many features of their human counterparts. These genetically engineered mouse (GEM) strains have provided important insights into the cellular and molecular determinants that underlie mouse Nf1 optic glioma development, maintenance, and associated vision loss, with relevance by extension to human NF1-OPG disease. Herein, we review our current understanding of NF1-OPG pathobiology and describe the mechanisms responsible for tumor initiation, growth, and associated vision loss in Nf1 GEM models. We also discuss how Nf1 GEM and other preclinical models can be deployed to identify and evaluate molecularly targeted therapies for OPG, particularly as they pertain to future strategies aimed at preventing or improving tumor-associated vision loss in children with NF1.
Collapse
Affiliation(s)
- Morgan E Freret
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
29
|
Evaluation of racial disparities in pediatric optic pathway glioma incidence: Results from the Surveillance, Epidemiology, and End Results Program, 2000-2014. Cancer Epidemiol 2018; 54:90-94. [PMID: 29684801 DOI: 10.1016/j.canep.2018.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 04/06/2018] [Accepted: 04/07/2018] [Indexed: 11/24/2022]
Abstract
BACKGROUND Racial predilection to pediatric cancer exists; however optic pathway glioma (OPG) risk differences by race/ethnicity are undefined. We estimated differences in OPG incidence across racial/ethnic groups in a multi-state cancer surveillance registry in the United States. METHODS OPG data were obtained from the Surveillance, Epidemiology, and End Results (SEER-18) Program, 2000-2014. Race/ethnicity was categorized as: White; Black; Asian; Other; and Latino/a ("Spanish-Hispanic-Latino"). Latino/a included all races, while all other categories excluded those identified as Latino/a. Age-adjusted incidence rates and rate ratios (IRR) with 95% confidence intervals (CIs) were generated in SEER-STAT (v8.3.4). RESULTS Data on 709 OPG cases ages 0-19 were abstracted from SEER-18. Minority children experienced lower age-adjusted OPG incidence rates compared to White children (IRRBlack = 0.38, 95% CI: 0.28-0.50; IRRAsian = 0.41, 95% CI: 0.29-0.58; and IRRLatino/a = 0.39, 95% CI: 0.32-0.48). In subgroup analyses among the highest risk age categories (0-4, 5-9), minority children experienced lower incidence rates compared to White children. Specific patterns for Latinos/as also emerged. Latino/a children ages 0-4 experienced the lowest incidence rates of all racial/ethnic groups compared to Whites (0.24 per 100,000 person-years versus 0.66 per 100,000 person-years, respectively), whereas among those ages 5-9, Black and Asian children experienced the lowest incidence rates (0.08 per 100,000 person-years each). CONCLUSIONS Incidence of OPGs was highest among White children. This study represents one of the largest to assess differences in OPG susceptibility by race/ethnicity. These findings may inform future studies that seek to evaluate modifying factors for this pediatric tumor including tumorigenesis, treatment, outcome, and long-term late effects.
Collapse
|
30
|
Jones DTW, Kieran MW, Bouffet E, Alexandrescu S, Bandopadhayay P, Bornhorst M, Ellison D, Fangusaro J, Fisher MJ, Foreman N, Fouladi M, Hargrave D, Hawkins C, Jabado N, Massimino M, Mueller S, Perilongo G, Schouten van Meeteren AYN, Tabori U, Warren K, Waanders AJ, Walker D, Weiss W, Witt O, Wright K, Zhu Y, Bowers DC, Pfister SM, Packer RJ. Pediatric low-grade gliomas: next biologically driven steps. Neuro Oncol 2018; 20:160-173. [PMID: 29016845 PMCID: PMC5786244 DOI: 10.1093/neuonc/nox141] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Despite the fact that they are not typically life-threatening, low-grade gliomas (LGGs) remain a significant clinical challenge in pediatric neuro-oncology due to comorbidities associated with these tumors and/or their treatments, and their propensity to multiply recurs. LGGs, in total the most common brain tumors arising in childhood, can often become a chronic problem requiring decades of management. The Second International Consensus Conference on Pediatric Low-Grade Gliomas held in Padua, Italy in 2016 was convened in an attempt to advance the pace of translating biological discoveries on LGGs into meaningful clinical benefit. Topics discussed included: the implications of our growing biological understanding of the genomics underlying these tumors; the assessment of the model systems available; the implications of the molecular and histopathologic differences between adult and pediatric diffuse gliomas; and steps needed to expedite targeted therapy into late-stage clinical trials for newly diagnosed cases. Methods for the diagnostic assessment of alterations in the Ras/mitogen-activated protein kinase pathway, typical for these tumors, were also considered. While the overall tone was positive, with a consensus that progress is being and will continue to be made, the scale of the challenge presented by this complex group of tumors was also acknowledged. The conclusions and recommendations of the meeting panel are provided here as an outline of current thinking and a basis for further discussion.
Collapse
Affiliation(s)
- David T W Jones
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Hopp Children’s Cancer Center, Heidelberg, Germany
| | - Mark W Kieran
- Department of Medical Oncology, Brigham and Women’s Hospital, Harvard Medical School, and the Broad Institutem, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| | - Eric Bouffet
- Paediatric Neuro-Oncology Program, Research Institute, The Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Sanda Alexandrescu
- Department of Pathology, Harvard Medical School, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Pratiti Bandopadhayay
- Department of Medical Oncology, Brigham and Women’s Hospital, Harvard Medical School, and the Broad Institutem, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| | - Miriam Bornhorst
- Gilbert Family Neurofibromatosis Institute, Brain Tumor Institute, Children’s National Health System, Washington DC, USA
- Center for Cancer and Immunology Research, Children’s National Health System, Washington DC, USA
| | - David Ellison
- Department of Pathology and 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, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Michael J Fisher
- Department of Pediatric Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Nicholas Foreman
- Children’s Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| | - Maryam Fouladi
- Brain Tumor Center, Brain Tumor Translational Research and Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Darren Hargrave
- Neuro-oncology and Experimental Therapeutics, Great Ormond Street Hospital for Children, London, UK
| | - Cynthia Hawkins
- Division of Pathology, The Arthur and Sonia Labatt Brain Tumor Research Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Nada Jabado
- Department of Pediatrics, McGill University, Montreal, Quebec, Canada
| | - Maura Massimino
- Pediatric Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Sabine Mueller
- 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
- Paediatric Neuro-Oncology Program, Research Institute, The Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Katherine Warren
- Department of Medical Oncology, Brigham and Women’s Hospital, Harvard Medical School, and the Broad Institutem, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
- National Cancer Institute, Pediatric Oncology and Neuro-Oncology Branches, Bethesda, Maryland, USA
| | - Angela J Waanders
- Department of Pediatric Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - David Walker
- Children’s Brain Tumor Research Centre, QMC University of Nottingham, Nottingham, UK
| | - William Weiss
- Department of Neurology, Pediatrics, and Neurosurgery, University of California San Francisco, San Francisco, California, USA
| | - Olaf Witt
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Hopp Children’s Cancer Center, Heidelberg, Germany
| | | | - Yuan Zhu
- Gilbert Family Neurofibromatosis Institute, Brain Tumor Institute, Children’s National Health System, Washington DC, USA
| | - Daniel C Bowers
- Department of Pediatrics, UT Southwestern Medical School, Dallas, Texas, USA
| | - Stefan M Pfister
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Hopp Children’s Cancer Center, Heidelberg, Germany
| | - Roger J Packer
- Gilbert Family Neurofibromatosis Institute, Brain Tumor Institute, Children’s National Health System, Washington DC, USA
- Center for Neuroscience and Behavioral Medicine, Gilbert Family Neurofibromatosis Institute, Brain Tumor Institute, Children’s National Health System, Washington DC, USA
| |
Collapse
|
31
|
Shofty B, Ben-Sira L, Kesler A, Jallo G, Groves ML, Iyer RR, Lassaletta A, Tabori U, Bouffet E, Thomale UW, Hernáiz Driever P, Constantini S. Isolated optic nerve gliomas: a multicenter historical cohort study. J Neurosurg Pediatr 2017; 20:549-555. [PMID: 28984541 DOI: 10.3171/2017.6.peds17107] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Isolated optic nerve gliomas (IONGs) constitute a rare subgroup of optic pathway gliomas (OPGs). Due to the rarity of this condition and the difficulty in differentiating IONGs from other types of OPGs in most clinical series, little is known about these tumors. Currently, due to lack of evidence, they are managed the same as any other OPG. METHODS The authors conducted a multicenter retrospective cohort study aimed at determining the natural history of IONGs. Included were patients with clear-cut glioma of the optic nerve without posterior (chiasmatic/hypothalamic) involvement. At least 1 year of follow-up, 2 MRI studies, and 2 neuro-ophthalmological examinations were required for inclusion. RESULTS Thirty-six patients with 39 tumors were included in this study. Age at diagnosis ranged between 6 months and 16 years (average 6 years). The mean follow-up time was 5.6 years. Twenty-five patients had neurofibromatosis Type 1. During the follow-up period, 59% of the tumors progressed, 23% remained stable, and 18% (all with neurofibromatosis Type 1) displayed some degree of spontaneous regression. Fifty-one percent of the patients presented with visual decline, of whom 90% experienced further deterioration. Nine patients were treated with chemotherapy, 5 of whom improved visually. Ten patients underwent operation, and no local or distal recurrence was noted. CONCLUSIONS Isolated optic nerve gliomas are highly dynamic tumors. Radiological progression and visual deterioration occur in greater percentages than in the general population of patients with OPGs. Response to chemotherapy may be better in this group, and its use should be considered early in the course of the disease.
Collapse
Affiliation(s)
- Ben Shofty
- 1The Gilbert Israeli Neurofibromatosis Center, Dana Children's Hospital.,2Division of Neurosurgery
| | - Liat Ben-Sira
- 1The Gilbert Israeli Neurofibromatosis Center, Dana Children's Hospital.,3Pediatric Radiology
| | - Anat Kesler
- 1The Gilbert Israeli Neurofibromatosis Center, Dana Children's Hospital.,4Division of Ophthalmology; and
| | - George Jallo
- 5Department of Neurosurgery, Johns Hopkins School of Medicine and Hospital, Baltimore, Maryland
| | - Mari L Groves
- 5Department of Neurosurgery, Johns Hopkins School of Medicine and Hospital, Baltimore, Maryland
| | - Rajiv R Iyer
- 5Department of Neurosurgery, Johns Hopkins School of Medicine and Hospital, Baltimore, Maryland
| | - Alvaro Lassaletta
- 6Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada; and
| | - Uri Tabori
- 6Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada; and
| | - Eric Bouffet
- 6Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada; and
| | - Ulrich-Wilhelm Thomale
- 7Pediatric Neurosurgery, Charité Universitätsmedizin, Campus Virchow Klinikum, Berlin, Germany
| | - Pablo Hernáiz Driever
- 7Pediatric Neurosurgery, Charité Universitätsmedizin, Campus Virchow Klinikum, Berlin, Germany
| | - Shlomi Constantini
- 1The Gilbert Israeli Neurofibromatosis Center, Dana Children's Hospital.,8Pediatric Neurosurgery, Dana Children's Hospital, Tel Aviv Medical Center and Tel Aviv University, Tel Aviv, Israel
| | | |
Collapse
|
32
|
Optic Pathway Gliomas in Neurofibromatosis Type 1: An Update: Surveillance, Treatment Indications, and Biomarkers of Vision. J Neuroophthalmol 2017; 37 Suppl 1:S23-S32. [PMID: 28806346 DOI: 10.1097/wno.0000000000000550] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Optic pathway gliomas (OPGs) occur in 15%-20% of children with neurofibromatosis type 1 (NF1), leading to visual deficits in fewer than half of these individuals. The goal of chemotherapy is to preserve vision, but vision loss in NF1-associated OPG can be unpredictable. Determining which child would benefit from chemotherapy and, equally important, which child is better observed without treatment can be difficult. Unfortunately, despite frequent imaging and ophthalmologic evaluations, some children experience progressive vision loss before treatment. Indications for chemotherapy usually are based on a comprehensive, quantitative assessment of vision, but reliable vision evaluation can be challenging in young children with NF1-OPG. The ability to identify and predict impending vision loss could potentially improve management decisions and visual outcomes. To address this challenge, ophthalmologic, electrophysiologic, and imaging biomarkers of vision in NF1-OPG have been proposed. We review current recommendations for the surveillance of children at risk for NF1-OPG, outline guidelines for initiating therapy, and describe the utility of proposed biomarkers for vision.
Collapse
|
33
|
Hales PW, Smith V, Dhanoa-Hayre D, O'Hare P, Mankad K, d'Arco F, Cooper J, Kaur R, Phipps K, Bowman R, Hargrave D, Clark C. Delineation of the visual pathway in paediatric optic pathway glioma patients using probabilistic tractography, and correlations with visual acuity. NEUROIMAGE-CLINICAL 2017. [PMID: 29527480 PMCID: PMC5842647 DOI: 10.1016/j.nicl.2017.10.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Background Radiological biomarkers which correlate with visual function are needed to improve the clinical management of optic pathway glioma (OPG) patients. Currently, these are not available using conventional magnetic resonance imaging (MRI) sequences. The aim of this study was to determine whether diffusion MRI could be used to delineate the entire optic pathway in OPG patients, and provide imaging biomarkers within this pathway which correlate with a patient's visual acuity (VA). Methods Multi-shell diffusion MRI data were acquired in a cohort of paediatric OPG patients, along with VA measurements in each eye. Diffusion MRI data were processed using constrained spherical deconvolution and probabilistic fibre tractography, to delineate the white matter bundles forming the optic pathway in each patient. Median fractional anisotropy (FA) and apparent diffusion coefficient (ADC) were measured in the optic nerves, tracts, and radiations, and correlated against each patient's VA. Results In the optic nerves, median FA significantly correlated with VA (R2adj = 0.31, p = 0.0082), with lower FA associated with poorer vision. In the optic radiations, both lower FA and higher ADC were significantly associated with poorer vision (R2adj = 0.52, p = 0.00075 and R2adj = 0.50, p = 0.0012 respectively). No significant correlations between VA and either FA or ADC were found in the optic tracts. Conclusions Multi-shell diffusion MRI provides in vivo delineation of the optic pathway in OPG patients, despite the presence of tumour invasion. This technique provides imaging biomarkers which are sensitive to microstructural damage to the underlying white matter in this pathway, which is not always visible on conventional MRI. Diffusion MRI can delineate the entire visual pathway in optic pathway glioma patients. Decreased FA in the optic nerves and radiations is associated with poorer vision. This provides sub-clinical biomarkers of structural damage to the visual pathway. These biomarkers correlate strongly with a patient's visual acuity.
Collapse
Affiliation(s)
- Patrick W Hales
- Developmental Imaging & Biophysics Section, University College London Great Ormond Street Institute of Child Health, London, UK.
| | - Victoria Smith
- Ophthalmology Department, Great Ormond Street Children's Hospital, London, UK
| | - Deepi Dhanoa-Hayre
- Ophthalmology Department, Great Ormond Street Children's Hospital, London, UK
| | - Patricia O'Hare
- Haematology and Oncology Department, Great Ormond Street Children's Hospital, London, UK
| | - Kshitij Mankad
- Radiology Department, Great Ormond Street Children's Hospital, London, UK
| | - Felice d'Arco
- Radiology Department, Great Ormond Street Children's Hospital, London, UK
| | - Jessica Cooper
- Radiology Department, Great Ormond Street Children's Hospital, London, UK
| | - Ramneek Kaur
- Developmental Imaging & Biophysics Section, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Kim Phipps
- Haematology and Oncology Department, Great Ormond Street Children's Hospital, London, UK
| | - Richard Bowman
- Ophthalmology Department, Great Ormond Street Children's Hospital, London, UK
| | - Darren Hargrave
- Haematology and Oncology Department, Great Ormond Street Children's Hospital, London, UK
| | - Christopher Clark
- Developmental Imaging & Biophysics Section, University College London Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
34
|
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
|
35
|
Pan Y, Bush EC, Toonen JA, Ma Y, Solga AC, Sims PA, Gutmann DH. Whole tumor RNA-sequencing and deconvolution reveal a clinically-prognostic PTEN/PI3K-regulated glioma transcriptional signature. Oncotarget 2017; 8:52474-52487. [PMID: 28881745 PMCID: PMC5581044 DOI: 10.18632/oncotarget.17193] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 03/30/2017] [Indexed: 12/29/2022] Open
Abstract
The concept that solid tumors are maintained by a productive interplay between neoplastic and non-neoplastic elements has gained traction with the demonstration that stromal fibroblasts and immune system cells dictate cancer development and progression. While less studied, brain tumor (glioma) biology is likewise influenced by non-neoplastic immune system cells (macrophages and microglia) which interact with neoplastic glioma cells to create a unique physiological state (glioma ecosystem) distinct from that found in the normal tissue. To explore this neoplastic ground state, we leveraged several preclinical mouse models of neurofibromatosis type 1 (NF1) optic glioma, a low-grade astrocytoma whose formation and maintenance requires productive interactions between non-neoplastic and neoplastic cells, and employed whole tumor RNA-sequencing and mathematical deconvolution strategies to characterize this low-grade glioma ecosystem as an aggregate of cellular and acellular elements. Using this approach, we demonstrate that optic gliomas generated by altering the germline Nf1 gene mutation, the glioma cell of origin, or the presence of co-existing genetic alterations represent molecularly-distinct tumors. However, these optic glioma tumors share a 25-gene core signature, not found in normal optic nerve, that is normalized by microglia inhibition (minocycline), but not conventional (carboplatin) or molecularly-targeted (rapamycin) chemotherapy. Lastly, we identify a genetic signature conferred by Pten reduction and corrected by PI3K inhibition. This signature predicts progression-free survival in patients with either low-grade or high-grade glioma. Collectively, these findings support the concept that gliomas are composite ecological systems whose biology and response to therapy may be best defined by examining the tumor as a whole.
Collapse
Affiliation(s)
- Yuan Pan
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Erin C Bush
- Departments of Systems Biology and of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, NY, USA
| | - Joseph A Toonen
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yu Ma
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne C Solga
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Peter A Sims
- Departments of Systems Biology and of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, NY, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
36
|
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: 2.7] [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
|
37
|
Borghei-Razavi H, Shibao S, Schick U. Prechiasmatic transection of the optic nerve in optic nerve glioma: technical description and surgical outcome. Neurosurg Rev 2016; 40:135-141. [PMID: 27230830 DOI: 10.1007/s10143-016-0747-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 03/11/2016] [Accepted: 05/05/2016] [Indexed: 11/28/2022]
Abstract
Optic pathway glioma (OPG) encompasses a spectrum of findings ranging from lesions confined to the optic nerve only, lesions affecting the optic chiasm and hypothalamus, and lesions with diffuse involvement of a large part of the optic pathway and neighboring structures. The majority of pediatric low-grade astrocytomas in the optic/chiasmatic region are typical pilocytic astrocytoma. The rest of them (10 %) may be other gliomas such as fibrillary pilomyxoid astrocytoma (grade 2 WHO). The postsurgical local recurrence rate of 55 to 76 % has been reported in some histological subtypes such as pilomyxoid astrocytoma (grade 2). Performing a prechiasmatic transection might offer a new surgical option to avoid further tumor growth toward the chiasm in the optic nerve glioma with predominantly orbital manifestations. In this retrospective study, four patients (three children, two without neurofibromatosis type 1 (NF1), and one with NF1 and one adult without NF1) with optic nerve glioma without involvement of the chiasm but blindness, disfiguring proptosis, and pain of the affected eye were included. The surgical approach was performed as a combined approach from pterional extradural and intradural. Without any exceptions, vision of the contralateral eye could be preserved and did not show any deterioration after surgery or during the follow-up time between 17 and 106 months. Furthermore, in all patients, gross total tumor resection could be achieved. During follow-up observation in all patients, no further tumor progress or recurrences could be observed. None of the patients were treated postoperatively by radiotherapy or chemotherapy. Prechiasmatic transection of the optic nerve in optic nerve glioma without affecting the chiasm might offer a surgical treatment option to control tumor growth and to preserve vision of the contralateral eye.
Collapse
Affiliation(s)
- Hamid Borghei-Razavi
- Department of Neurosurgery, Clemens Hospital, Academic Hospital of Münster University, Düesbergweg 124, 48153, Münster, Germany.
| | - Shunsuke Shibao
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Uta Schick
- Department of Neurosurgery, Clemens Hospital, Academic Hospital of Münster University, Düesbergweg 124, 48153, Münster, Germany
| |
Collapse
|
38
|
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: 8.1] [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
|
39
|
Abstract
Two children without neurofibromatosis type 1 presented with unilateral decreased vision and MRI revealing optic nerve tumors. In the first case, chemotherapy was initiated empirically for presumed optic pathway glioma, but the lesion increased in size with associated clinical worsening, raising concern for a possible alternate diagnosis. Biopsy of the involved optic nerve resulted in worsening of vision due to a branch retinal artery occlusion and showed a grade I pilocytic astrocytoma. In the second case, sudden symptom onset and rapid tumor growth prompted an optic nerve biopsy, resulting in vision loss due to a central retinal artery occlusion and revealing grade I pilocytic astrocytoma. In both cases, tissue diagnosis did not alter the course of management. Instead, biopsy was associated with additional vision loss, highlighting the risk of biopsy in children with isolated optic nerve tumors and imaging that is most consistent with an optic pathway glioma.
Collapse
|
40
|
Falsini B, Chiaretti A, Rizzo D, Piccardi M, Ruggiero A, Manni L, Soligo M, Dickmann A, Federici M, Salerni A, Timelli L, Guglielmi G, Lazzareschi I, Caldarelli M, Galli-Resta L, Colosimo C, Riccardi R. Nerve growth factor improves visual loss in childhood optic gliomas: a randomized, double-blind, phase II clinical trial. Brain 2016; 139:404-414. [PMID: 26767384 DOI: 10.1093/brain/awv366] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/26/2015] [Indexed: 11/15/2022] Open
Abstract
Paediatric optic pathway gliomas are low-grade brain tumours characterized by slow progression and invalidating visual loss. Presently there is no strategy to prevent visual loss in this kind of tumour. This study evaluated the effects of nerve growth factor administration in protecting visual function in patients with optic pathway glioma-related visual impairment. A prospective randomized double-blind phase II clinical trial was conducted in 18 optic pathway glioma patients, aged from 2 to 23 years, with stable disease and severe visual loss. Ten patients were randomly assigned to receive a single 10-day course of 0.5 mg murine nerve growth factor as eye drops, while eight patients received placebo. All patients were evaluated before and after treatment, testing visual acuity, visual field, visual-evoked potentials, optic coherence tomography, electroretinographic photopic negative response, and magnetic resonance imaging. Post-treatment evaluations were repeated at 15, 30, 90, and 180 days Brain magnetic resonance imaging was performed at baseline and at 180 days. Treatment with nerve growth factor led to statistically significant improvements in objective electrophysiological parameters (electroretinographic photopic negative response amplitude at 180 days and visual-evoked potentials at 30 days), which were not observed in placebo-treated patients. Furthermore, in patients in whom visual fields could still be measured, visual field worsening was only observed in placebo-treated cases, while three of four nerve growth factor-treated subjects showed significant visual field enlargement. This corresponded to improved visually guided behaviour, as reported by the patients and/or the caregivers. There was no evidence of side effects related to nerve growth factor treatment. Nerve growth factor eye drop administration appears a safe, easy and effective strategy for the treatment of visual loss associated with optic pathway gliomas.
Collapse
Affiliation(s)
- Benedetto Falsini
- 1 Institute of Ophthalmology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonio Chiaretti
- 2 Paediatric Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Daniela Rizzo
- 2 Paediatric Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Marco Piccardi
- 1 Institute of Ophthalmology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonio Ruggiero
- 2 Paediatric Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Luigi Manni
- 3 Institute of Translational Pharmacology, CNR, 00142 Rome, Italy
| | - Marzia Soligo
- 3 Institute of Translational Pharmacology, CNR, 00142 Rome, Italy
| | - Anna Dickmann
- 1 Institute of Ophthalmology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Matteo Federici
- 1 Institute of Ophthalmology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Annabella Salerni
- 1 Institute of Ophthalmology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | - Gaspare Guglielmi
- 5 Pharmacy Gemelli Hospital, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Ilaria Lazzareschi
- 2 Paediatric Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Massimo Caldarelli
- 6 Paediatric Neurosurgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | - Cesare Colosimo
- 8 Institute of Radiology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Riccardo Riccardi
- 2 Paediatric Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
41
|
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: 2.7] [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
|
42
|
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
|
43
|
Shofty B, Mauda-Havakuk M, Weizman L, Constantini S, Ben-Bashat D, Dvir R, Pratt LT, Joskowicz L, Kesler A, Yalon M, Ravid L, Ben-Sira L. The effect of chemotherapy on optic pathway gliomas and their sub-components: A volumetric MR analysis study. Pediatr Blood Cancer 2015; 62:1353-9. [PMID: 25858021 DOI: 10.1002/pbc.25480] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 01/26/2015] [Indexed: 01/03/2023]
Abstract
BACKGROUND Optic pathway gliomas (OPG) represent 5% of pediatric brain tumors and compose a major therapeutic dilemma to the treating physicians. While chemotherapy is widely used for these tumors, our ability to predict radiological response is still lacking. In this study, we use volumetric imaging to examine in detail the long-term effect of chemotherapy on the tumor as well as its various sub-components. PROCEDURE The tumors of 15 patients with OPG, treated with chemotherapy, were longitudinally measured using our novel, previously described volumetric method. Patients were treated with up to five lines of chemotherapy. Sufficient follow-up imaging data, and patient's numbers, allowed for analysis of two treatment lines. Volumetric measurements of the tumors were segmented into solid-non-enhancing, solid-enhancing, and cystic components. Outcome analysis was done per specific treatment line and for the overall follow-up period. RESULTS An average reduction of 9.7% (±23%) in the gross-total-solid volume (GTSV) was noted following treatment with vincristine and carboplatin. The cystic component grew under therapy by an average of 12.6% (±39%). When measured over the course of the whole study period, the cystic component grew by an average of 35% (±100%) and the GTSV increased by 12% (±35%). CONCLUSION Initial treatment with vincristine and carboplatin seems to have a minimal initial effect, mostly on the solid components. The cystic component in itself seems to be unaffected by chemotherapy, and contributes to the subsequent growth of the total volume. During the overall treatment period, both solid and cystic components grew regardless of combined treatment methods.
Collapse
Affiliation(s)
- Ben Shofty
- The Gilbert Israeli Neurofibromatosis Center.,Pediatric Neurosurgery Dana Children's Hospital
| | | | - Lior Weizman
- Functional Brain Center, The Wohl Institute for Advanced Imaging
| | - Shlomi Constantini
- The Gilbert Israeli Neurofibromatosis Center.,Pediatric Neurosurgery Dana Children's Hospital
| | - Dafna Ben-Bashat
- Pediatric Hematology-Oncology, all at the Tel-Aviv Medical Center and The Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Rina Dvir
- School of Engineering and Computer Science, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Li-Tal Pratt
- The Gilbert Israeli Neurofibromatosis Center.,Pediatric Radiology Unit
| | - Leo Joskowicz
- Functional Brain Center, The Wohl Institute for Advanced Imaging
| | - Anat Kesler
- The Gilbert Israeli Neurofibromatosis Center
| | - Michal Yalon
- Pediatric Hematology Oncology, Sheba Medical Center, Tel-Hashomer, Israel
| | - Lior Ravid
- The Gilbert Israeli Neurofibromatosis Center
| | - Liat Ben-Sira
- The Gilbert Israeli Neurofibromatosis Center.,Pediatric Radiology Unit
| |
Collapse
|
44
|
Abstract
Optic pathway gliomas (OPGs) are among the most challenging neoplasms in modern pediatric neuro-oncology. Recent technological advances in imaging, surgery, and chemotherapy may lead to better understanding of the pathophysiology and better clinical results. This chapter reviews these advances and the current treatment paradigms.
Collapse
Affiliation(s)
- Ben Shofty
- Department of Pediatric Neurosurgery, Dana Children's Hospital, Tel-Aviv Medical Center, Tel Aviv University, 6th Weizmann St., Tel-Aviv, 64239, Israel
| | | | | | | |
Collapse
|
45
|
Shofty B, Constantini S, Bokstein F, Ram Z, Ben-Sira L, Freedman S, Vainer G, Kesler A. Optic pathway gliomas in adults. Neurosurgery 2014; 74:273-9; discussion 279-80. [PMID: 24335817 DOI: 10.1227/neu.0000000000000257] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Optic pathway gliomas (OPGs) are considered relatively benign pediatric tumors. Adult patients with OPG can be divided into 2 groups: adult patients with tumors diagnosed in childhood and adult patients diagnosed during adulthood. OBJECTIVE To characterize the clinical course of adult patients with OPG. METHODS We retrospectively collected clinical and imaging data of all adult OPG patients monitored in our medical center between 1990 and 2012. RESULTS Twenty-two adult patients were included. Age at diagnosis varied widely (6 months-66 years), as did age at last follow-up (18-74 years). Ten patients were diagnosed at adulthood and 12 in childhood. Of the patients diagnosed at childhood, 6 had radiological progression during childhood, and 3 of those patients suffered visual impairment. From this group, 1 patient had further progression during adulthood accompanied by additional visual decline, and 2 patients had additional visual decline during adulthood despite no signs of progression. Of the 6 patients whose tumors were stable during childhood, all 6 remained stable during adulthood. Of 10 patients diagnosed at adulthood, 6 patients suffered visual deterioration; in 5 of them, a concomitant progression was noted. Two patients were diagnosed with high-grade gliomas. CONCLUSION OPGs may be active during childhood or adulthood. Those patients who experienced anatomic activity during childhood are prone to continue experiencing active disease during adulthood. A significant percentage of patients diagnosed with low-grade OPG at adulthood may suffer progression, visual decline, or both. ABBREVIATIONS NF1, neurofibromatosis 1OPG, optic pathway gliomas.
Collapse
Affiliation(s)
- Ben Shofty
- *Division of Neurosurgery, ‡Gilbert Israeli Neurofibromatosis Center, §Neuro-Oncology Service, ¶Pediatric Radiology Unit, ‖Pathology, and #Neuro-Ophthalmology Unit, Tel-Aviv Medical Center, and Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Kalin-Hajdu E, Décarie JC, Marzouki M, Carret AS, Ospina LH. Visual acuity of children treated with chemotherapy for optic pathway gliomas. Pediatr Blood Cancer 2014; 61:223-7. [PMID: 23956233 DOI: 10.1002/pbc.24726] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 07/16/2013] [Indexed: 01/05/2023]
Abstract
BACKGROUND Chemotherapy is the most common primary treatment modality for pediatric optic pathway gliomas (OPGs). Due to the risk of severe visual impairment, visual acuity (VA) has become a clinical parameter of fundamental importance for children with OPGs. Despite this reality, most studies omit crucial information necessary for analysis of the effect of chemotherapy on VA in patients with cerebral gliomas. The principal goal of this study was to determine the immediate and long-term visual outcome of children treated first with chemotherapy for OPGs. PROCEDURE Retrospective, non-comparative, case series of children with OPGs treated initially with chemotherapy. VA was measured prior to chemotherapy, directly following chemotherapy, as well as at last follow-up. RESULTS Seven children (14 eyes) were positive for the neurofibromatosis type-1 (NF1) mutation and 10 children (20 eyes) were without the NF1 mutation (sporadic). Three deaths, all in the sporadic cohort, occurred as a result of their OPG. Median follow-up time of survivors was 10.54 ± 4.36 (SD) years. Both NF1 mutation positive and sporadic cohorts had deterioration in VA over time; however, deterioration was only statistically significant in the sporadic population. The percentage of eyes with vision weaker than 20/200 prior to chemotherapy, directly following chemotherapy and at last follow-up grew from 18% to 24% to 38%, respectively. CONCLUSIONS In both NF1 mutant and sporadic OPGs, VA deteriorated directly following chemotherapy as well as at long-term follow-up. Despite chemotherapy, eyes with severe functional impairment gradually increased over time.
Collapse
Affiliation(s)
- Evan Kalin-Hajdu
- CHU Sainte Justine, University of Montreal, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
47
|
Fisher MJ, Avery RA, Allen JC, Ardern-Holmes SL, Bilaniuk LT, Ferner RE, Gutmann DH, Listernick R, Martin S, Ullrich NJ, Liu GT, REiNS International Collaboration. Functional outcome measures for NF1-associated optic pathway glioma clinical trials. Neurology 2013; 81:S15-24. [PMID: 24249802 PMCID: PMC3908337 DOI: 10.1212/01.wnl.0000435745.95155.b8] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 08/13/2013] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE The goal of the Response Evaluation in Neurofibromatosis and Schwannomatosis Visual Outcomes Committee is to define the best functional outcome measures for future neurofibromatosis type 1 (NF1)-associated optic pathway glioma (OPG) clinical trials. METHODS The committee considered the components of vision, other ophthalmologic parameters affected by OPG, potential biomarkers of visual function, and quality of life measures to arrive at consensus-based, evidence-driven recommendations for objective and measurable functional endpoints for OPG trials. RESULTS Visual acuity (VA) assessments using consistent quantitative testing methods are recommended as the main functional outcome measure for NF1-OPG clinical trials. Teller acuity cards are recommended for use as the primary VA endpoint, and HOTV as a secondary endpoint once subjects are old enough to complete it. The optic disc should be assessed for pallor, as this appears to be a contributory variable that may affect the interpretation of VA change over time. Given the importance of capturing patient-reported outcomes in clinical trials, evaluating visual quality of life using the Children's Visual Function Questionnaire as a secondary endpoint is also proposed. CONCLUSIONS The use of these key functional endpoints will be essential for evaluating the efficacy of future OPG clinical trials.
Collapse
Affiliation(s)
- Michael J Fisher
- From the Division of Oncology (M.J.F.), Neuroradiology Section, Department of Radiology (L.T.B.), and Neuro-Ophthalmology Service (G.T.L.), The Children's Hospital of Philadelphia; Department of Pediatrics (M.J.F.) and Departments of Neurology and Ophthalmology (G.T.L.), The Perelman School of Medicine at the University of Pennsylvania, Philadelphia; Departments of Neurology, Ophthalmology, and Pediatrics (R.A.A.), Gilbert Family Neurofibromatosis Institute, Children's National Medical Center, Washington, DC; Departments of Pediatrics and Neurology (J.C.A.), NYU Cancer Institute, NYU Langone Medical Center, New York, NY; Children's Hospital at Westmead Clinical School (S.L.A.-H.), The University of Sydney, Australia; Department of Neurology (S.L.A.-H.), The Children's Hospital at Westmead, Sydney, Australia; University of Pennsylvania School of Medicine (L.T.B.), Philadelphia; Department of Neurology (R.E.F.), Guy's and St. Thomas' NHS Foundation Trust and Institute of Psychiatry, King's College London; Department of Neurology (D.H.G.), Washington University School of Medicine, St. Louis, MO; Department of Pediatrics (R.L.), Feinberg School of Medicine, Northwestern University; Ann & Robert H. Lurie Children's Hospital of Chicago (R.L.); Pediatric Oncology Branch (S.M.), National Cancer Institute, National Institutes of Health, Bethesda, MD; and Department of Neurology (N.J.U.), Boston Children's Hospital, Harvard Medical School, Boston, MA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Treatment of neuro-ophthalmic and orbitofacial manifestations of neurofibromatosis type 1. Curr Opin Ophthalmol 2013; 24:506-11. [DOI: 10.1097/icu.0b013e32836348a4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
49
|
de Blank PMK, Berman JI, Liu GT, Roberts TPL, Fisher MJ. Fractional anisotropy of the optic radiations is associated with visual acuity loss in optic pathway gliomas of neurofibromatosis type 1. Neuro Oncol 2013; 15:1088-95. [PMID: 23658320 DOI: 10.1093/neuonc/not068] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND No more than half of patients with neurofibromatosis type 1 (NF1)-associated optic pathway gliomas (OPGs) develop vision loss. Prospectively identifying those who will require therapy remains challenging, because no reliable factors have yet been identified that predict future vision loss. To determine whether brain tissue microstructure is associated with visual acuity loss, we examined diffusion tensor imaging (DTI) and ophthalmologic evaluations in children with NF1-associated OPG. METHODS We retrospectively reviewed ophthalmology records and concurrent DTI measurements of the optic nerves, tracts, and radiations from 50 children with NF1-associated OPGs. Multivariate linear regression measured the association between fiber trajectory quantity and white matter integrity on visual acuity measured by the logarithm of the minimal angle of resolution (logMAR). RESULTS In multivariate analysis, fractional anisotropy (FA) of the optic radiations was associated with visual acuity loss (adjusted coefficient = -6.081 logMAR/FA; P = .006) after adjusting for age, extent of tumor, DTI acquisition type, prior chemotherapy, and fundus examination findings. The association remained after eliminating tumors involving the optic radiations. In an evaluation of 15 subjects with paired ophthalmologic examination and DTI a year apart, initial FA of the optic radiation was associated with a trend toward change in visual acuity a year later (coefficient = -2.652 logMAR/FA; P = .069). CONCLUSIONS A decrease in FA of the optic radiations is associated with abnormal visual acuity in NF1-associated OPGs and may be predictive of visual acuity loss during the following year.
Collapse
Affiliation(s)
- Peter Matthew Kennedy de Blank
- Division of Pediatric Hematology and Oncology, Rainbow Babies and Children’s Hospital and Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA.
| | | | | | | | | |
Collapse
|
50
|
Terashima K, Chow K, Jones J, Ahern C, Jo E, Ellezam B, Paulino AC, Okcu MF, Su J, Adesina A, Mahajan A, Dauser R, Whitehead W, Lau C, Chintagumpala M. Long-term outcome of centrally located low-grade glioma in children. Cancer 2013; 119:2630-8. [PMID: 23625612 DOI: 10.1002/cncr.28110] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 03/05/2013] [Accepted: 03/06/2013] [Indexed: 11/07/2022]
Abstract
BACKGROUND Optimal management of children with centrally located low-grade glioma (LGG) is unclear. Initial interventions in most children are chemotherapy in younger and radiation therapy (RT) in older children. A better understanding of the inherent risk factors along with the effects of interventions on long-term outcome can lead to reassessment of the current approaches to minimize long-term morbidity. METHODS To reassess the current treatment strategies of centrally located LGG, we compared the long-term survival and morbidity of different treatment regimens. Medical records of patients primarily treated at Texas Children's Cancer and Hematology Centers between 1987 and 2008 were reviewed. RESULTS Forty-seven patients with a median follow-up of 79 months were included in the analysis. The 5-year overall survival and progression-free survival (PFS) for all patients were 96% and 53%, respectively. The 5-year PFS for those treated initially with RT (12 patients; median age, 11 years [range, 3-15 years]) and with chemotherapy (28 patients; median age, 2 years [range 0-8 years]) were 76% and 37%, respectively (log-rank test P = .02). Among children who progressed after chemotherapy, the 5-year PFS after salvage RT was 55%. Patients diagnosed at a younger age (<5 years) were more likely to experience endocrine abnormalities (Fisher exact test; P<.00001). CONCLUSIONS Effective and durable tumor control was obtained with RT as initial treatment. In younger patients, chemotherapy can delay the use of RT; however, frequent progression and long-term morbidity are common. More effective and less toxic therapies are required in these patients, the majority of whom are long-term survivors.
Collapse
Affiliation(s)
- Keita Terashima
- Texas Children's Cancer and Hematology Centers, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|