1
|
Thomson N, Jewels E, Bergeron-Gravel S, Lagacé S, Hazrati LN, Saikali S, Dunham C, Cheng S. YAP1-MAML2 Fusion in Young Children with Pediatric High-Grade Glioma: A Case Report. Pediatr Blood Cancer 2025:e31796. [PMID: 40394848 DOI: 10.1002/pbc.31796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2025] [Accepted: 05/03/2025] [Indexed: 05/22/2025]
Affiliation(s)
- Nina Thomson
- Division of Hematology, Oncology & BMT, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Emily Jewels
- Division of Hematology, Oncology & BMT, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Samuel Bergeron-Gravel
- Department of Radiation Oncology, CHU de Québec, Université Laval, Québec, Québec, Canada
| | - Simon Lagacé
- Department of Radiation Oncology, CHU de Québec, Université Laval, Québec, Québec, Canada
| | - Lili-Naz Hazrati
- Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Québec, Québec, Canada
| | - Stephen Saikali
- Department of Pathology, CHU de Québec, Université Laval, Québec, Québec, Canada
| | - Christopher Dunham
- Division of Anatomical Pathology, BC Children's Hospital and Women's Hospital and Health Center, Vancouver, British Columbia, Canada
| | - Sylvia Cheng
- Division of Hematology, Oncology & BMT, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
2
|
McThenia SS, Reddy KM, Damaraju E, Castellino E, He Z, Beers R, Chien F, Castellino RC, Goldman-Yassen AE, Fangusaro JR, MacDonald T. BRAF inhibitor monotherapy in BRAFV600E-mutated pediatric low-grade glioma: a single center's experience. Front Oncol 2025; 14:1505951. [PMID: 39839763 PMCID: PMC11747024 DOI: 10.3389/fonc.2024.1505951] [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: 10/10/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
Background Pediatric low-grade gliomas (pLGGs) have an overall survival of over 90%; however, patients harboring a BRAFV600E alteration may have worse outcomes, particularly when treated with classic chemotherapy. Combined BRAF/MEK inhibition following incomplete resection demonstrated improved outcome in BRAFV600E altered pLGG compared to combined carboplatin/vincristine chemotherapy and is now considered the standard FDA-approved treatment for this group of tumors. The aim herein was to investigate the efficacy and tolerability of single agent BRAF inhibitor treatment in BRAFV600E altered pLGG. Methods A single institution retrospective chart review analysis was performed on patients, 0 to 21 years of age, with newly diagnosed and/or progressive BRAFV600E mutated pLGGs (WHO Grade 1 or 2) at Children's Healthcare of Atlanta treated off-study with BRAF inhibitor monotherapy between 2013-2023. 2-year progression free survival (PFS) and objective tumor response was evaluated. All toxicities possibly associated with BRAF inhibition therapy were evaluated and described according to Common Terminology Criteria for Adverse Events version 5 (CTCAEv5). MRI brain imaging data at baseline and best response was evaluated to identify patterns that may predict response to BRAF inhibition monotherapy. Results Fifteen patients diagnosed with BRAFV600E mutated pLGG, treated with monotherapy BRAF inhibition, were identified. Median age of diagnosis: 3.8 years (0.2 -18.1). Histologic diagnosis: pilocytic astrocytoma (PA) (N=4); ganglioglioma (GGL) (N=3); GGL, atypical (N=3); pleomorphic xanthroastrocytoma (PXA) (N=2); low-grade neuroepithelial tumor (N=1); infiltrating glioma (N=1); and LGG (NOS) (N=1). Tumor locations included: hypothalamus/optic chiasm (N=6); brainstem (N=4); third ventricle/thalamus (N=2); parietal/temporal lobe (N=2); and spinal cord (N=1). Mean duration of BRAF inhibitor monotherapy: 38.41 months (range 3.9-83.7). Median follow-up: 32.6 months (16 - 78.1). Two-year PFS for patients on BRAFi monotherapy for at least 10 months: 90% (95% CI: 73.2%-100%). Objective Response (OR) for 15 evaluable patients on BRAF inhibitor (BRAFi) therapy: 73% (0/15 CR + 6/15 PR + 5/15 MR) with Overall Response Rate (ORR=CR+PR): 40%. Overall, patients tolerated treatment well with Grade 1 rash being the most common toxicity. Two of 15 patients (13%) discontinued therapy due to toxicities, and 2 other patients switched within drug class from vemurafenib to dabrafenib due to toxicities. Discussion In this small cohort of incompletely resected BRAFV600E mutated pLGGs, BRAFi monotherapy was effective and well tolerated with an ORR comparable to published prospective outcomes of dual MEK/BRAF inhibitor therapy. This promising monotherapy treatment should be considered when choosing treatment for incompletely resected BRAFV600E-altered pLGGs.
Collapse
Affiliation(s)
- S. S. McThenia
- Department of Pediatrics, Children’s Healthcare of Atlanta, Atlanta, GA, United States
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Aflac Cancer & Blood Disorders Center, Atlanta, GA, United States
| | - K. M. Reddy
- Department of Radiology, Children’s Healthcare of Atlanta, Atlanta, GA, United States
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, United States
| | - E. Damaraju
- Department of Radiology, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - E. Castellino
- Department of Radiology, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Z. He
- Department of Pediatrics, Emory University, Atlanta, GA, United States
| | - R. Beers
- Department of Pediatrics, Emory University, Atlanta, GA, United States
| | - F. Chien
- Department of Pediatrics, Children’s Healthcare of Atlanta, Atlanta, GA, United States
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Aflac Cancer & Blood Disorders Center, Atlanta, GA, United States
| | - R. C. Castellino
- Department of Pediatrics, Children’s Healthcare of Atlanta, Atlanta, GA, United States
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Aflac Cancer & Blood Disorders Center, Atlanta, GA, United States
| | - A. E. Goldman-Yassen
- Department of Radiology, Children’s Healthcare of Atlanta, Atlanta, GA, United States
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, United States
| | - J. R. Fangusaro
- Department of Pediatrics, Children’s Healthcare of Atlanta, Atlanta, GA, United States
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Aflac Cancer & Blood Disorders Center, Atlanta, GA, United States
| | - T. MacDonald
- Department of Pediatrics, Children’s Healthcare of Atlanta, Atlanta, GA, United States
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Aflac Cancer & Blood Disorders Center, Atlanta, GA, United States
| |
Collapse
|
3
|
Geraldo AF, Maldonado F, Severino M, Mankad K, Dahmoush H, Soares B, Rugilo C, Rossi A. Response assessment in pediatric neurooncology (RAPNO) criteria revisited: a practical navigation guide for neuroradiologists. Neuroradiology 2024; 66:2117-2142. [PMID: 39446196 DOI: 10.1007/s00234-024-03493-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024]
Abstract
The Response Assessment in Pediatric Neuro-Oncology (RAPNO) Working Group is an international, collaborative network of experts dedicated to pediatric central nervous system (CNS) tumors that was created in 2011. Since then, six RAPNO articles with imaging guidelines for response assessment in diverse pediatric tumor subgroups have been published, namely: 1) medulloblastomas and leptomeningeal seeding tumors (2018), 2) pediatric high-grade gliomas (2020), 3) pediatric low-grade gliomas (2020), 4) diffuse intrinsic pontine gliomas (2020), 5) pediatric intracranial ependymomas (2022) and 6) pediatric craniopharyngiomas (2023). The purpose of this article is to review all current available RAPNO criteria using a systematized and comparative approach centered on the role of neuroradiologists and supported by neuroimaging examples. Special emphasis will be placed on clarification of core concepts as well as practical adoption aspects of the RAPNO guidelines, namely how and when to image the brain and/or the spine; how to interpret the imaging findings; which other clinical, therapeutic and laboratory variables to consider; and finally how to apply the information to attribute the final appropriate response assessment classification.
Collapse
Affiliation(s)
- Ana Filipa Geraldo
- Diagnostic Neuroradiology Unit, Department of Radiology, Unidade Local de Saúde Gaia/Espinho (ULSGE), Vila Nova de Gaia, Portugal.
- Department of Clinical Neurosciences and Mental Health, Faculty of Medicine of the University of Porto, Porto, Portugal.
| | - Francisco Maldonado
- Diagnostic Neuroradiology Unit, Department of Radiology, Hospital de Pediatría Prof Dr Juan P Garrahan, Buenos Aires, Argentina
| | | | - Kshitij Mankad
- Department of Radiology, Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Hisham Dahmoush
- Department of Radiology, Lucile Packard Children's Hospital at Stanford, Palo Alto, CA, USA
| | - Bruno Soares
- Department of Radiology, Lucile Packard Children's Hospital at Stanford, Palo Alto, CA, USA
| | - Carlos Rugilo
- Diagnostic Neuroradiology Unit, Department of Radiology, Hospital de Pediatría Prof Dr Juan P Garrahan, Buenos Aires, Argentina
| | - Andrea Rossi
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| |
Collapse
|
4
|
Familiar AM, Fathi Kazerooni A, Vossough A, Ware JB, Bagheri S, Khalili N, Anderson H, Haldar D, Storm PB, Resnick AC, Kann BH, Aboian M, Kline C, Weller M, Huang RY, Chang SM, Fangusaro JR, Hoffman LM, Mueller S, Prados M, Nabavizadeh A. Towards consistency in pediatric brain tumor measurements: Challenges, solutions, and the role of artificial intelligence-based segmentation. Neuro Oncol 2024; 26:1557-1571. [PMID: 38769022 PMCID: PMC11376457 DOI: 10.1093/neuonc/noae093] [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: 02/13/2024] [Indexed: 05/22/2024] Open
Abstract
MR imaging is central to the assessment of tumor burden and changes over time in neuro-oncology. Several response assessment guidelines have been set forth by the Response Assessment in Pediatric Neuro-Oncology (RAPNO) working groups in different tumor histologies; however, the visual delineation of tumor components using MRIs is not always straightforward, and complexities not currently addressed by these criteria can introduce inter- and intra-observer variability in manual assessments. Differentiation of non-enhancing tumors from peritumoral edema, mild enhancement from absence of enhancement, and various cystic components can be challenging; particularly given a lack of sufficient and uniform imaging protocols in clinical practice. Automated tumor segmentation with artificial intelligence (AI) may be able to provide more objective delineations, but rely on accurate and consistent training data created manually (ground truth). Herein, this paper reviews existing challenges and potential solutions to identifying and defining subregions of pediatric brain tumors (PBTs) that are not explicitly addressed by current guidelines. The goal is to assert the importance of defining and adopting criteria for addressing these challenges, as it will be critical to achieving standardized tumor measurements and reproducible response assessment in PBTs, ultimately leading to more precise outcome metrics and accurate comparisons among clinical studies.
Collapse
Affiliation(s)
- Ariana M Familiar
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Anahita Fathi Kazerooni
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Arastoo Vossough
- Division of Radiology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jeffrey B Ware
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sina Bagheri
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nastaran Khalili
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Hannah Anderson
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Debanjan Haldar
- Department of Neurosurgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Phillip B Storm
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Adam C Resnick
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Benjamin H Kann
- Department of Radiation Oncology, Dana-Farber Cancer Institute | Brigham and Women’s Hospital | Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mariam Aboian
- Division of Radiology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Cassie Kline
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Susan M Chang
- Division of Neuro-Oncology, Department of Neurosurgery, University of California, San Francisco, California, USA
| | - Jason R Fangusaro
- The Aflac Cancer Center, Children’s Healthcare of Atlanta and the Emory University School of Medicine, Atlanta, Georgia, USA
| | - Lindsey M Hoffman
- Division of Hematology/Oncology, Phoenix Children’s Hospital, Phoenix, Arizona, USA
| | - Sabine Mueller
- Department of Neurology, Neurosurgery and Pediatrics, University of California, San Francisco, California, USA
| | - Michael Prados
- Department of Neurosurgery and Pediatrics, University of California, San Francisco, California, USA
| | - Ali Nabavizadeh
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
5
|
Vossough A, Khalili N, Familiar AM, Gandhi D, Viswanathan K, Tu W, Haldar D, Bagheri S, Anderson H, Haldar S, Storm PB, Resnick A, Ware JB, Nabavizadeh A, Fathi Kazerooni A. Training and Comparison of nnU-Net and DeepMedic Methods for Autosegmentation of Pediatric Brain Tumors. AJNR Am J Neuroradiol 2024; 45:1081-1089. [PMID: 38724204 PMCID: PMC11383404 DOI: 10.3174/ajnr.a8293] [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: 01/05/2024] [Accepted: 03/01/2024] [Indexed: 08/11/2024]
Abstract
BACKGROUND AND PURPOSE Tumor segmentation is essential in surgical and treatment planning and response assessment and monitoring in pediatric brain tumors, the leading cause of cancer-related death among children. However, manual segmentation is time-consuming and has high interoperator variability, underscoring the need for more efficient methods. After training, we compared 2 deep-learning-based 3D segmentation models, DeepMedic and nnU-Net, with pediatric-specific multi-institutional brain tumor data based on multiparametric MR images. MATERIALS AND METHODS Multiparametric preoperative MR imaging scans of 339 pediatric patients (n = 293 internal and n = 46 external cohorts) with a variety of tumor subtypes were preprocessed and manually segmented into 4 tumor subregions, ie, enhancing tumor, nonenhancing tumor, cystic components, and peritumoral edema. After training, performances of the 2 models on internal and external test sets were evaluated with reference to ground truth manual segmentations. Additionally, concordance was assessed by comparing the volume of the subregions as a percentage of the whole tumor between model predictions and ground truth segmentations using the Pearson or Spearman correlation coefficients and the Bland-Altman method. RESULTS The mean Dice score for nnU-Net internal test set was 0.9 (SD, 0.07) (median, 0.94) for whole tumor; 0.77 (SD, 0.29) for enhancing tumor; 0.66 (SD, 0.32) for nonenhancing tumor; 0.71 (SD, 0.33) for cystic components, and 0.71 (SD, 0.40) for peritumoral edema, respectively. For DeepMedic, the mean Dice scores were 0.82 (SD, 0.16) for whole tumor; 0.66 (SD, 0.32) for enhancing tumor; 0.48 (SD, 0.27) for nonenhancing tumor; 0.48 (SD, 0.36) for cystic components, and 0.19 (SD, 0.33) for peritumoral edema, respectively. Dice scores were significantly higher for nnU-Net (P ≤ .01). Correlation coefficients for tumor subregion percentage volumes were higher (0.98 versus 0.91 for enhancing tumor, 0.97 versus 0.75 for nonenhancing tumor, 0.98 versus 0.80 for cystic components, 0.95 versus 0.33 for peritumoral edema in the internal test set). Bland-Altman plots were better for nnU-Net compared with DeepMedic. External validation of the trained nnU-Net model on the multi-institutional Brain Tumor Segmentation Challenge in Pediatrics (BraTS-PEDs) 2023 data set revealed high generalization capability in the segmentation of whole tumor, tumor core (a combination of enhancing tumor, nonenhancing tumor, and cystic components), and enhancing tumor with mean Dice scores of 0.87 (SD, 0.13) (median, 0.91), 0.83 (SD, 0.18) (median, 0.89), and 0.48 (SD, 0.38) (median, 0.58), respectively. CONCLUSIONS The pediatric-specific data-trained nnU-Net model is superior to DeepMedic for whole tumor and subregion segmentation of pediatric brain tumors.
Collapse
Affiliation(s)
- Arastoo Vossough
- From the Center for Data Driven Discovery in Biomedicine (A.V., N.K., A.M.F., D.G., K.V., D.H., S.B., H.A., P.B.S., A.R., A.N., A.F.K.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Radiology (A.V., S.B., J.B.W., A.N.), University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Radiology (A.V.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Nastaran Khalili
- From the Center for Data Driven Discovery in Biomedicine (A.V., N.K., A.M.F., D.G., K.V., D.H., S.B., H.A., P.B.S., A.R., A.N., A.F.K.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Ariana M Familiar
- From the Center for Data Driven Discovery in Biomedicine (A.V., N.K., A.M.F., D.G., K.V., D.H., S.B., H.A., P.B.S., A.R., A.N., A.F.K.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Deep Gandhi
- From the Center for Data Driven Discovery in Biomedicine (A.V., N.K., A.M.F., D.G., K.V., D.H., S.B., H.A., P.B.S., A.R., A.N., A.F.K.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Karthik Viswanathan
- From the Center for Data Driven Discovery in Biomedicine (A.V., N.K., A.M.F., D.G., K.V., D.H., S.B., H.A., P.B.S., A.R., A.N., A.F.K.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Wenxin Tu
- College of Arts and Sciences (W.T.), University of Pennsylvania, Philadelphia, Pennsylvania
| | - Debanjan Haldar
- From the Center for Data Driven Discovery in Biomedicine (A.V., N.K., A.M.F., D.G., K.V., D.H., S.B., H.A., P.B.S., A.R., A.N., A.F.K.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Sina Bagheri
- From the Center for Data Driven Discovery in Biomedicine (A.V., N.K., A.M.F., D.G., K.V., D.H., S.B., H.A., P.B.S., A.R., A.N., A.F.K.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Radiology (A.V., S.B., J.B.W., A.N.), University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hannah Anderson
- From the Center for Data Driven Discovery in Biomedicine (A.V., N.K., A.M.F., D.G., K.V., D.H., S.B., H.A., P.B.S., A.R., A.N., A.F.K.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Shuvanjan Haldar
- School of Engineering (S.H.), Rutgers University, New Brunswick, New Jersey
| | - Phillip B Storm
- From the Center for Data Driven Discovery in Biomedicine (A.V., N.K., A.M.F., D.G., K.V., D.H., S.B., H.A., P.B.S., A.R., A.N., A.F.K.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Neurosurgery (P.B.S., A.F.K.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Adam Resnick
- From the Center for Data Driven Discovery in Biomedicine (A.V., N.K., A.M.F., D.G., K.V., D.H., S.B., H.A., P.B.S., A.R., A.N., A.F.K.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Jeffrey B Ware
- Department of Radiology (A.V., S.B., J.B.W., A.N.), University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ali Nabavizadeh
- From the Center for Data Driven Discovery in Biomedicine (A.V., N.K., A.M.F., D.G., K.V., D.H., S.B., H.A., P.B.S., A.R., A.N., A.F.K.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Radiology (A.V., S.B., J.B.W., A.N.), University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anahita Fathi Kazerooni
- From the Center for Data Driven Discovery in Biomedicine (A.V., N.K., A.M.F., D.G., K.V., D.H., S.B., H.A., P.B.S., A.R., A.N., A.F.K.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Neurosurgery (P.B.S., A.F.K.), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Center for AI & Data Science for Integrated Diagnostics (A.F.K.), University of Pennsylvania, Philadelphia, Pennsylvania
- Center for Biomedical Image Computing and Analytics (A.F.K.), University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
6
|
Yu X, Lai M, Li J, Wang L, Ye K, Zhang D, Hu Q, Li S, Hu X, Wang Q, Ma M, Xiao Z, Zhou J, Shi C, Luo L, Cai L. The relationship between imaging features, therapeutic response, and overall survival in pediatric diffuse intrinsic pontine glioma. Neurosurg Rev 2024; 47:212. [PMID: 38727935 PMCID: PMC11087318 DOI: 10.1007/s10143-024-02435-8] [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: 11/29/2023] [Revised: 04/24/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024]
Abstract
We aimed to evaluate the relationship between imaging features, therapeutic responses (comparative cross-product and volumetric measurements), and overall survival (OS) in pediatric diffuse intrinsic pontine glioma (DIPG). A total of 134 patients (≤ 18 years) diagnosed with DIPG were included. Univariate and multivariate analyses were performed to evaluate correlations of clinical and imaging features and therapeutic responses with OS. The correlation between cross-product (CP) and volume thresholds in partial response (PR) was evaluated by linear regression. The log-rank test was used to compare OS patients with discordant therapeutic response classifications and those with concordant classifications. In univariate analysis, characteristics related to worse OS included lower Karnofsky, larger extrapontine extension, ring-enhancement, necrosis, non-PR, and increased ring enhancement post-radiotherapy. In the multivariate analysis, Karnofsky, necrosis, extrapontine extension, and therapeutic response can predict OS. A 25% CP reduction (PR) correlated with a 32% volume reduction (R2 = 0.888). Eight patients had discordant therapeutic response classifications according to CP (25%) and volume (32%). This eight patients' median survival time was 13.0 months, significantly higher than that in the non-PR group (8.9 months), in which responses were consistently classified as non-PR based on CP (25%) and volume (32%). We identified correlations between imaging features, therapeutic responses, and OS; this information is crucial for future clinical trials. Tumor volume may represent the DIPG growth pattern more accurately than CP measurement and can be used to evaluate therapeutic response.
Collapse
Affiliation(s)
- Xiaojun Yu
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Mingyao Lai
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China
| | - Juan Li
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China
| | - Lichao Wang
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China
| | - Kunlin Ye
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Dong Zhang
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Qingjun Hu
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China
| | - Shaoqun Li
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China
| | - Xinpeng Hu
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Qiong Wang
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Mengjie Ma
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Zeyu Xiao
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Jiangfen Zhou
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China
| | - Changzheng Shi
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China.
| | - Liangping Luo
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China.
- Department of Medical Imaging Center, The Fifth Affiliated Hospital of Jinan University, Yingke Avenue, Heyuan City, 517000, China.
| | - Linbo Cai
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China.
| |
Collapse
|
7
|
Tang ER, Alazraki AL, Thacker PG, McCarville MB, Towbin AJ. Introduction to the COG Diagnostic Imaging Committee/SPR Oncology Committee White Papers: Rationale and methods. Pediatr Blood Cancer 2023; 70 Suppl 4:e30411. [PMID: 37158569 PMCID: PMC10626871 DOI: 10.1002/pbc.30411] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/10/2023]
Abstract
Pediatric cancer is a rare disease. Because of this, many sites do not have experience providing imaging for specific tumor types. The Children's Oncology Group Diagnostic Imaging Committee and the Society for Pediatric Radiology Oncology Committee are comprised of radiologists with expertise in pediatric cancer imaging. Recently, this group endeavored to create a series of 23 White Papers designed to provide evidence-based imaging recommendations and minimum achievable imaging protocols. The purpose of this manuscript is to describe the methods employed in authoring the White Paper series.
Collapse
Affiliation(s)
- Elizabeth R. Tang
- Department of Radiology, Section of Pediatric Radiology, Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Adina L. Alazraki
- Departments of Pediatrics and Radiology, Emory University School of Medicine, Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Paul G. Thacker
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - M Beth McCarville
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Alexander J. Towbin
- Department of Radiology, Cincinnati Children’s Hospital; Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
8
|
Laudicella R, Mantarro C, Catalfamo B, Alongi P, Gaeta M, Minutoli F, Baldari S, Bisdas S. PET Imaging in Gliomas. RADIOLOGY‐NUCLEAR MEDICINE DIAGNOSTIC IMAGING 2023:194-218. [DOI: 10.1002/9781119603627.ch6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
9
|
Ramakrishnan D, von Reppert M, Krycia M, Sala M, Mueller S, Aneja S, Nabavizadeh A, Galldiks N, Lohmann P, Raji C, Ikuta I, Memon F, Weinberg BD, Aboian MS. Evolution and implementation of radiographic response criteria in neuro-oncology. Neurooncol Adv 2023; 5:vdad118. [PMID: 37860269 PMCID: PMC10584081 DOI: 10.1093/noajnl/vdad118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
Radiographic response assessment in neuro-oncology is critical in clinical practice and trials. Conventional criteria, such as the MacDonald and response assessment in neuro-oncology (RANO) criteria, rely on bidimensional (2D) measurements of a single tumor cross-section. Although RANO criteria are established for response assessment in clinical trials, there is a critical need to address the complexity of brain tumor treatment response with multiple new approaches being proposed. These include volumetric analysis of tumor compartments, structured MRI reporting systems like the Brain Tumor Reporting and Data System, and standardized approaches to advanced imaging techniques to distinguish tumor response from treatment effects. In this review, we discuss the strengths and limitations of different neuro-oncology response criteria and summarize current research findings on the role of novel response methods in neuro-oncology clinical trials and practice.
Collapse
Affiliation(s)
- Divya Ramakrishnan
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Marc von Reppert
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Mark Krycia
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Matthew Sala
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Sabine Mueller
- Department of Neurology, Neurosurgery, and Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Sanjay Aneja
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Ali Nabavizadeh
- Department of Radiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-3), Research Center Juelich, Juelich, Germany
- Center for Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Duesseldorf, Cologne, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-4), Research Center Juelich, Juelich, Germany
| | - Cyrus Raji
- Department of Radiology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Ichiro Ikuta
- Department of Radiology, Mayo Clinic, Phoenix, Arizona, USA
| | - Fatima Memon
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Brent D Weinberg
- Department of Radiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mariam S Aboian
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
10
|
Lindsay HB, Massimino M, Avula S, Stivaros S, Grundy R, Metrock K, Bhatia A, Fernández-Teijeiro A, Chiapparini L, Bennett J, Wright K, Hoffman LM, Smith A, Pajtler KW, Poussaint TY, Warren KE, Foreman NK, Mirsky DM. Response assessment in paediatric intracranial ependymoma: recommendations from the Response Assessment in Pediatric Neuro-Oncology (RAPNO) working group. Lancet Oncol 2022; 23:e393-e401. [DOI: 10.1016/s1470-2045(22)00222-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 10/16/2022]
|
11
|
Lazow MA, Nievelstein MT, Lane A, Bandopadhayhay P, DeWire-Schottmiller M, Fouladi M, Glod JW, Greiner RJ, Hoffman LM, Hummel TR, Kilburn L, Leary S, Minturn JE, Packer R, Ziegler DS, Chaney B, Black K, de Blank P, Leach JL. Volumetric endpoints in diffuse intrinsic pontine glioma: comparison to cross-sectional measures and outcome correlations in the International DIPG/DMG Registry. Neuro Oncol 2022; 24:1598-1608. [PMID: 35148393 PMCID: PMC9435485 DOI: 10.1093/neuonc/noac037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Cross-sectional tumor measures are traditional clinical trial endpoints; however volumetric measures may better assess tumor growth. We determined the correlation and compared the prognostic impact of cross-sectional and volumetric measures of progressive disease (PD) among patients with DIPG. METHODS Imaging and clinical data were abstracted from the International DIPG Registry. Tumor volume and cross-sectional product (CP) were measured with mint Lesion™ software using manual contouring. Correlation between CP and volume (segmented and mathematical [ellipsoid] model) thresholds of PD were assessed by linear regression. Landmark analyses determined differences in survival (via log-rank) between patients classified as PD versus non-PD by CP and volumetric measurements at 1, 3, 5, 7, and 9 months postradiotherapy (RT). Hazard ratios (HR) for survival after these time points were calculated by Cox regression. RESULTS A total of 312 MRIs (46 patients) were analyzed. Comparing change from the previous smallest measure, CP increase of 25% (PD) correlated with a segmented volume increase of 30% (R2 = 0.710), rather than 40% (spherical model extrapolation). CP-determined PD predicted survival at 1 month post-RT (HR = 2.77), but not other time points. Segmented volumetric-determined PD (40% threshold) predicted survival at all imaging timepoints (HRs = 2.57, 2.62, 3.35, 2.71, 16.29), and 30% volumetric PD threshold predicted survival at 1, 3, 5, and 9 month timepoints (HRs = 2.57, 2.62, 4.65, 5.54). Compared to ellipsoid volume, segmented volume demonstrated superior survival associations. CONCLUSIONS Segmented volumetric assessments of PD correlated better with survival than CP or ellipsoid volume at most time points. Semiautomated tumor volume likely represents a more accurate, prognostically-relevant measure of disease burden in DIPG.
Collapse
Affiliation(s)
| | | | - Adam Lane
- Brain Tumor Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | | | - Maryam Fouladi
- Pediatric Neuro-Oncology Program, Nationwide Children’s Hospital, Columbus, Ohio, USA,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - John W Glod
- Cancer for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Robert J Greiner
- Division of Oncology, Penn State Health Children’s Hospital, Hershey, Pennsylvania, USA
| | - Lindsey M Hoffman
- Division of Oncology, Phoenix Children’s Hospital, Phoenix, Arizona, USA
| | - Trent R Hummel
- Brain Tumor Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Lindsay Kilburn
- Division of Oncology, Children’s National Medical Center, Washington, DC, USA
| | - Sarah Leary
- Cancer and Blood Disorders Center, Seattle Children’s Hospital, Seattle, Washington, USA
| | - Jane E Minturn
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Roger Packer
- Division of Oncology, Children’s National Medical Center, Washington, DC, USA
| | - David S Ziegler
- 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
| | - Brooklyn Chaney
- Brain Tumor Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Katie Black
- Brain Tumor Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | | | - James L Leach
- Corresponding Author: James L. Leach, MD, Department of Radiology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Avenue Cincinnati, OH 45229, USA ()
| |
Collapse
|
12
|
Abstract
Clinical trials play a critical role in discovering new treatments, but the path to regulatory approval can be cumbersome and time consuming. Efforts to increase the efficiency and interpretability of clinical trials within the neuro-oncology community have focused on standardization of response assessment, development of consensus guidelines for clinical trial conduct, decentralization of clinical trials, removal of barriers to clinical trial accrual, and re-examination of patient eligibility criteria.
Collapse
Affiliation(s)
- Eudocia Q Lee
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
| |
Collapse
|
13
|
Gnekow AK, Kandels D, Pietsch T, Bison B, Warmuth-Metz M, Thomale UW, Kortmann RD, Timmermann B, Driever PH, Witt O, Schmidt R, Spix C. Doubling Recruitment of Pediatric Low-grade Glioma within Two Decades does not change Outcome - Report from the German LGG Studies. KLINISCHE PADIATRIE 2021; 233:107-122. [PMID: 33971689 DOI: 10.1055/a-1471-5897] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Successive multicenter studies for pediatric low-grade glioma (LGG) in Germany were accompanied by a doubling of annual recruitment over 2 decades. We investigated whether this increase conveyed a change of epidemiologic characteristics or survival. METHODS AND RESULTS Participating centers reported 4634 patients with the radiologic/histologic diagnosis of LGG (1996-2018), rising from 109 to 278/year. Relating these numbers to all pediatric CNS tumors registered at the German Childhood Cancer Registry, the LGG fraction and annual crude incidence rates increased (32% to 51%; 0.94 to 2.12/100,000 children/adolescents<15 years). The consecutive LGG studies recruited 899 (HIT-LGG 1996), 1592 (SIOP-LGG 2004), and 1836 (LGG-registry) patients with similar distribution of tumor-sites, histology, and dissemination. 5-year overall survival was 96%-98% at median observation time of 8.1 years. Acknowledging unequal follow-up periods, 589/899 (66%), 1089/1582 (69%), and 1387/1836 (76%) patients remained under observation, while 1252/4317 received adjuvant treatment with decreasing frequency of front-line radiotherapy from 16% to 5%. CONCLUSION Pediatric LGG incidence rates in Germany are now comparable to other European countries. The rise in patient numbers followed implementation of standard-of-care treatment protocols, but did not result in relevant changes of epidemiologic or clinical parameters or survival. Shifts in patient distribution between treatment arms reflect growing acceptance of the LGG therapy algorithm. HINTERGRUND In den vergangenen 20 Jahren hat sich die jährliche Patientenrekrutierung in den aufeinanderfolgenden multizentrischen Studien für pädiatrische niedrig-gradige Gliome (LGG) in Deutschland verdoppelt. Wir haben untersucht, ob sich mit dieser Zunahme auch epidemiologische Merkmale oder das Überleben verändert haben. METHODIK UND ERGEBNISSE Zwischen 1996 und 2018 meldeten die teilnehmenden Zentren insgesamt 4634 Patienten mit der radiologischen/histologischen Diagnose eines LGG. Die Zahl stieg von anfangs 109 bis 278 Patienten pro Jahr. Gleichzeitig stieg der Anteil der LGGs an allen am Deutschen Kinderkrebsregister gemeldeten pädiatrischen Hirntumoren von 32 auf 51%, die jährliche Inzidenz erhöhte sich von 0,94 auf 2,12/100 000 Kinder/Jugendliche<15 Jahre. Die aufeinanderfolgenden LGG-Studien rekrutierten 899 (HIT-LGG 1996), 1592 (SIOP-LGG 2004) und 1836 (LGG-Register) Patienten mit vergleichbarer Verteilung von Tumorsitz, Histologie und Disseminierung. Das 5-Jahres-Überleben lag bei einer medianen Nachbeobachtungszeit von 8,1 Jahren zwischen 96 und 98%. Unter Berücksichtigung der ungleich langen Follow-up-Zeit wurden 589/899 (65,5%), 1089/1582 (68,8%) und 1387/1836 (75,5%) Patienten bislang beobachtet, während 1252/4317 eine adjuvante Therapie erhielten. Dabei sank der Anteil der primären Radiotherapie von 16 auf 5%. SCHLUSSFOLGERUNG Die Rekrutierung pädiatrischer LGG ist dank Implementierung verbindlicher Therapiestandards in Deutschland gestiegen, ohne zu relevanten Veränderungen epidemiologischer oder klinischer Merkmale oder des Überlebens zu führen. Die Inzidenz ist mit anderen europäischen Ländern vergleichbar. Verschiebungen der Patientenzuteilung zwischen den Therapiearmen spiegeln die zunehmende Akzeptanz des LGG-Therapie-Algorithmus wider.
Collapse
Affiliation(s)
- Astrid Katharina Gnekow
- Faculty of Medicine, Swabian Children's Cancer Center, University Hospital Augsburg, Augsburg, Germany
| | - Daniela Kandels
- Faculty of Medicine, Swabian Children's Cancer Center, University Hospital Augsburg, Augsburg, Germany
| | - Torsten Pietsch
- Institute of Neuropathology, DGNN Brain Tumor Reference Center, University Bonn, Bonn, Germany
| | - Brigitte Bison
- Institute of Diagnostic and Interventional Neuroradiology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Monika Warmuth-Metz
- Institute of Diagnostic and Interventional Neuroradiology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Ulrich W Thomale
- Department of Pediatric Neuro-Surgery, Charité Berlin, Berlin, Germany
| | - Rolf-D Kortmann
- Referenzzentrum für Hirntumoren (Radioonkologie), Universitätsklinikum Leipzig AöR, Leipzig, Germany
| | - Beate Timmermann
- Westgerman Proton Therapy Center Essen, University Essen, Essen, Germany
| | - Pablo Hernàiz Driever
- Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Olaf Witt
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - René Schmidt
- Institute of Biostatistics and Clinical Research, University of Muenster, Muenster, Germany
| | - Claudia Spix
- Division of Childhood Cancer Epidemiology, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
14
|
Peng J, Zhou H, Tang O, Chang K, Wang P, Zeng X, Shen Q, Wu J, Xiao Y, Patel SH, Hu C, Jin K, Xiao B, Boxerman J, Gao X, Wen PY, Bai HX, Huang RY, Yang L. Evaluation of RAPNO criteria in medulloblastoma and other leptomeningeal seeding tumors using MRI and clinical data. Neuro Oncol 2021; 22:1536-1544. [PMID: 32215549 DOI: 10.1093/neuonc/noaa072] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Although the Response Assessment in Pediatric Neuro-Oncology (RAPNO) working group has made recommendations for response assessment in patients with medulloblastoma (MBL) and leptomeningeal seeding tumors, these criteria have yet to be evaluated. METHODS We examined MR imaging and clinical data in a multicenter retrospective cohort of 269 patients with MBL diagnoses, high grade glioma, embryonal tumor, germ cell tumor, or choroid plexus papilloma. Interobserver agreement, objective response (OR) rates, and progression-free survival (PFS) were calculated. Landmark analyses were performed for OR and progression status at 0.5, 1.0, and 1.5 years after treatment initiation. Cox proportional hazards models were used to determine the associations between OR and progression with overall survival (OS). Subgroup analyses based on tumor subgroup and treatment modality were performed. RESULTS The median follow-up time was 4.0 years. In all patients, the OR rate was .0.565 (95% CI: 0.505-0.625) by RAPNO. The interobserver agreement of OR determination between 2 raters (a neuroradiologist and a neuro-oncologist) for the RAPNO criteria in all patients was 83.8% (k statistic = 0.815; P < 0.001). At 0.5-, 1.0-, and 1.5-year landmarks, both OR status and PFS determined by RAPNO were predictive of OS (hazard ratios [HRs] for 1-year landmark: OR HR = 0.079, P < 0.001; PFS HR = 10.192, P < 0.001). In subgroup analysis, OR status and PFS were predictive of OS for all tumor subtypes and treatment modalities. CONCLUSION RAPNO criteria showed excellent consistency in the treatment response evaluation of MBL and other leptomeningeal seeding tumors. OR and PFS determined by RAPNO criteria correlated with OS.
Collapse
Affiliation(s)
- Jian Peng
- Department of Neurology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hao Zhou
- Department of Neurology, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Oliver Tang
- Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Ken Chang
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Panpan Wang
- Department of Neurology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiaowei Zeng
- Department of Neurology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qin Shen
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jing Wu
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yanhe Xiao
- Department of Neurology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Sohil H Patel
- Department of Radiology and Medical Imaging, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Chongyu Hu
- Department of Neurology, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Ke Jin
- Department of Radiology, Hunan Children's Hospital, Changsha, Hunan, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jerrold Boxerman
- Department of Diagnostic Imaging, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Xiaoping Gao
- Department of Neurology, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Harrison X Bai
- Department of Diagnostic Imaging, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island, USA.,Department of Radiology, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Li Yang
- Department of Neurology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
15
|
Trunin YY, Golanov AV, Konovalov AN, Pronin IN, Zagirov RI, Ryzhova MV, Kadyrov SU, Igoshina EN. [Stereotactic irradiation in the complex treatment of patients with intracranial pilocytic astrocytoma]. ZHURNAL VOPROSY NEĬROKHIRURGII IMENI N. N. BURDENKO 2021; 85:34-46. [PMID: 33864667 DOI: 10.17116/neiro20218502134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Complex management of patients with intracranial pilocytic astrocytoma (PA) consists of surgical treatment, drug therapy (mainly in young children) and radiotherapy. For many years, radiotherapy (RT) has been a standard for residual tumors, recurrence or continued growth of PA. Currently, stereotactic radiosurgery and radiotherapy are preferred for PA, because these procedures are characterized by high conformity and selectivity, precise irradiation of tumor with minimal damage to surrounding intact tissues. Stereotaxic approach is very important since PAs are localized near functionally significant and radiosensitive brain structures in most cases. There is significant experience of single-center studies devoted to radiotherapy of patients with PA at the Department of Neuroradiosurgery of the Burdenko Neurosurgery Center. In this research, the authors analyzed the results of stereotactic irradiation of 430 patients with PA for the period from 2005 to 2018.
Collapse
Affiliation(s)
| | - A V Golanov
- Burdenko Neurosurgical Center, Moscow, Russia
| | | | - I N Pronin
- Burdenko Neurosurgical Center, Moscow, Russia
| | - R I Zagirov
- Burdenko Neurosurgical Center, Moscow, Russia
| | - M V Ryzhova
- Burdenko Neurosurgical Center, Moscow, Russia
| | | | | |
Collapse
|
16
|
Abstract
Primary pediatric brain tumors comprise a broad group of neoplasm subtypes that can be categorized based on their histological and molecular features according to the 2016 World Health Organization (WHO) classification of central nervous system (CNS) tumors. The majority of the pediatric brain tumors demonstrate a singular preference for this age group and have a unique molecular profile. The separation of certain tumor entities, including different types of embryonal tumors, low-grade gliomas, and high-grade gliomas, may have a significant impact by guiding appropriate treatment for these children and potentially changing their outcomes. Currently, the focus of the imaging diagnostic studies is to follow the molecular updates, searching for potential imaging patterns that translate this information in molecular profile results, therefore helping the final diagnosis. Due to the high impact of accurate diagnosis in this context, the scientific community has presented extensive research on imaging pediatric tumors in recent years. This article summarizes the key characteristics of the imaging features of the most common primary childhood brain tumors, categorizing them according to the recent WHO classification update, which is based on each of their molecular profiles. The purpose of this review article is to familiarize radiologists with their key imaging features and thereby improve diagnostic accuracy.
Collapse
|
17
|
Goldman S, Pollack IF, Jakacki RI, Billups CA, Poussaint TY, Adesina AM, Panigrahy A, Parsons DW, Broniscer A, Robinson GW, Robison NJ, Partap S, Kilburn LB, Onar-Thomas A, Dunkel IJ, Fouladi M. Phase II study of peginterferon alpha-2b for patients with unresectable or recurrent craniopharyngiomas: a Pediatric Brain Tumor Consortium report. Neuro Oncol 2021; 22:1696-1704. [PMID: 32393959 DOI: 10.1093/neuonc/noaa119] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Craniopharyngiomas account for approximately 1.2-4% of all CNS tumors. They are typically treated with a combination of surgical resection and focal radiotherapy. Unfortunately, treatment can lead to permanent deleterious effects on behavior, learning, and endocrine function. METHODS The Pediatric Brain Tumor Consortium performed a multicenter phase 2 study in children and young adults with unresectable or recurrent craniopharyngioma (PBTC-039). Between December 2013 and November 2017, nineteen patients (median age at enrollment, 13.1 y; range, 2-25 y) were enrolled in one of 2 strata: patients previously treated with surgery alone (stratum 1) or who received radiation (stratum 2). RESULTS Eighteen eligible patients (8 male, 10 female) were treated with weekly subcutaneous pegylated interferon alpha-2b for up to 18 courses (108 wk). Therapy was well tolerated with no grade 4 or 5 toxicities. 2 of the 7 eligible patients (28.6%) in stratum 1 had a partial response, but only one response was sustained for more than 3 months. None of the 11 stratum 2 patients had an objective radiographic response, although median progression-free survival was 19.5 months. CONCLUSIONS Pegylated interferon alpha-2b treatment, in lieu of or following radiotherapy, was well tolerated in children and young adults with recurrent craniopharyngiomas. Although objective responses were limited, progression-free survival results are encouraging, warranting further studies.
Collapse
Affiliation(s)
- Stewart Goldman
- Division of Hematology, Oncology, Neuro-Oncology, Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Ian F Pollack
- Department of Pediatric Neurosurgery, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Regina I Jakacki
- Department of Pediatric Neurosurgery, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Catherine A Billups
- Department of Biostatistics, St Jude's Children's Research Hospital, Memphis, Tennessee
| | - Tina Y Poussaint
- Department of Radiology, Boston Children's Hospital, Boston, Massachusetts.,Department of Radiology, Harvard Medical School, Boston, Massachusetts
| | | | - Ashok Panigrahy
- Department of Pediatric Radiology, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Donald W Parsons
- Texas Children's Cancer and Hematology Centers, Texas Medical Center, Houston, Texas
| | - Alberto Broniscer
- Department of Radiology, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Giles W Robinson
- Division of Neuro-Oncology, St Jude's Children's Research Hospital, Memphis, Tennessee
| | - Nathan J Robison
- Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, California
| | - Sonia Partap
- Department of Neurology, Stanford University School of Medicine, Stanford, California
| | - Lindsay B Kilburn
- Department of Hematology and Oncology, Children's National Medical Center, Washington, DC
| | - Arzu Onar-Thomas
- Department of Biostatistics, St Jude's Children's Research Hospital, Memphis, Tennessee
| | - Ira J Dunkel
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maryam Fouladi
- Department of Hematology and Oncology, Cincinnati Children's Hospital, Cincinnati, Ohio
| |
Collapse
|
18
|
Abstract
Malignant gliomas constitute a smaller portion of brain tumors in children compared with adults. Nevertheless, they can be devastating tumors with poor prognosis. Recent advances and improved understanding of the genetic and molecular characterization of pediatric brain tumors, including those of malignant gliomas, have led to the reclassification of many pediatric brain tumors and new entities have been defined. In this paper, we will present some of the more recent characterization and pertinent changes in pediatric high-grade gliomas, along with the conventional and advanced imaging features associated with these entities. Implications of the recent changes in pediatric malignant glioma classifications will also be discussed.
Collapse
|
19
|
Erker C, Tamrazi B, Poussaint TY, Mueller S, Mata-Mbemba D, Franceschi E, Brandes AA, Rao A, Haworth KB, Wen PY, Goldman S, Vezina G, MacDonald TJ, Dunkel IJ, Morgan PS, Jaspan T, Prados MD, Warren KE. Response assessment in paediatric high-grade glioma: recommendations from the Response Assessment in Pediatric Neuro-Oncology (RAPNO) working group. Lancet Oncol 2020; 21:e317-e329. [PMID: 32502458 DOI: 10.1016/s1470-2045(20)30173-x] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/05/2020] [Accepted: 03/12/2020] [Indexed: 12/27/2022]
Abstract
Response criteria for paediatric high-grade glioma vary historically and across different cooperative groups. The Response Assessment in Neuro-Oncology working group developed response criteria for adult high-grade glioma, but these were not created to meet the unique challenges in children with the disease. The Response Assessment in Pediatric Neuro-Oncology (RAPNO) working group, consisting of an international panel of paediatric and adult neuro-oncologists, clinicians, radiologists, radiation oncologists, and neurosurgeons, was established to address issues and unique challenges in assessing response in children with CNS tumours. We established a subcommittee to develop response assessment criteria for paediatric high-grade glioma. Current practice and literature were reviewed to identify major challenges in assessing the response of paediatric high-grade gliomas to various treatments. For areas in which scientific investigation was scarce, consensus was reached through an iterative process. RAPNO response assessment recommendations include the use of MRI of the brain and the spine, assessment of clinical status, and the use of corticosteroids or antiangiogenics. Imaging standards for brain and spine are defined. Compared with the recommendations for the management of adult high-grade glioma, for paediatrics there is inclusion of diffusion-weighted imaging and a higher reliance on T2-weighted fluid-attenuated inversion recovery. Consensus recommendations and response definitions have been established and, similar to other RAPNO recommendations, prospective validation in clinical trials is warranted.
Collapse
Affiliation(s)
- Craig Erker
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Dalhousie University and IWK Health Centre, Halifax, NS, Canada.
| | - Benita Tamrazi
- Department of Radiology, Keck School of Medicine, University of Southern California and Children's Hospital of Los Angeles, Los Angeles, CA, USA
| | - Tina Y Poussaint
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA
| | - Sabine Mueller
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA; Department of Neurosurgery, University of California San Francisco, San Francisco, CA, USA
| | - Daddy Mata-Mbemba
- Department of Diagnostic Imaging, Dalhousie University and IWK Health Centre, Halifax, NS, Canada
| | - Enrico Franceschi
- Department of Medical Oncology, Azienda USL, Bologna, Italy; IRCCS Institute of Neurological Sciences, Bologna, Italy
| | - Alba A Brandes
- Department of Medical Oncology, Azienda USL, Bologna, Italy; IRCCS Institute of Neurological Sciences, Bologna, Italy
| | - Arvind Rao
- Departments of Computational Medicine and Bioinformatics and Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Kellie B Haworth
- Division of Neuro-Oncology, Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Patrick Y Wen
- Center For Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Stewart Goldman
- Department of Haematology, Oncology, Neuro-Oncology, and Stem Cell Transplantation, Ann and Robert H Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Gilbert Vezina
- Department of Radiology, Children's National Medical Center, Washington, DC, USA
| | - Tobey J MacDonald
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Ira J Dunkel
- Department of Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paul S Morgan
- Department of Medical Physics and Clinical Engineering, Nottingham University Hospitals, Queen's Medical Centre, Nottingham, UK
| | - Tim Jaspan
- Department of Radiology, Nottingham University Hospitals, Queen's Medical Centre, Nottingham, UK
| | - Michael D Prados
- Department of Neurosurgery, University of California San Francisco, San Francisco, CA, USA
| | - Katherine E Warren
- Department of Pediatric Oncology, Dana- Farber/Boston Children's Cancer and Blood Disorders Center, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
20
|
Cooney TM, Cohen KJ, Guimaraes CV, Dhall G, Leach J, Massimino M, Erbetta A, Chiapparini L, Malbari F, Kramer K, Pollack IF, Baxter P, Laughlin S, Patay Z, Young Poussaint T, Warren KE. Response assessment in diffuse intrinsic pontine glioma: recommendations from the Response Assessment in Pediatric Neuro-Oncology (RAPNO) working group. Lancet Oncol 2020; 21:e330-e336. [PMID: 32502459 DOI: 10.1016/s1470-2045(20)30166-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/25/2020] [Accepted: 03/04/2020] [Indexed: 12/20/2022]
Abstract
Optimising the conduct of clinical trials for diffuse intrinsic pontine glioma involves use of consistent, objective disease assessments and standardised response criteria. The Response Assessment in Pediatric Neuro-Oncology working group, consisting of an international panel of paediatric and adult neuro-oncologists, clinicians, radiologists, radiation oncologists, and neurosurgeons, was established to address issues and unique challenges in assessing response in children with CNS tumours. A working group was formed specifically to address response assessment in children and young adults with diffuse intrinsic pontine glioma and to develop a consensus on recommendations for response assessment. Response should be assessed using MRI of brain and spine, neurological examination, and anti-inflammatory or antiangiogenic drugs. Clinical imaging standards are defined. As with previous consensus recommendations, these recommendations will need to be validated in prospective clinical trials.
Collapse
Affiliation(s)
- Tabitha M Cooney
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kenneth J Cohen
- Departments of Pediatrics and Oncology, Johns Hopkins University, Baltimore, MD, USA
| | | | - Girish Dhall
- Department of Pediatrics, Division of Hematology-Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - James Leach
- Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Maura Massimino
- Department of Pediatric Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessandra Erbetta
- Department of Neuroradiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Luisa Chiapparini
- Department of Neuroradiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Fatema Malbari
- Department of Pediatrics, Section of Neurology and Developmental Neurosciences, Texas Children's Hospital, Houston, TX, USA
| | - Kim Kramer
- Department of Pediatric Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ian F Pollack
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Patricia Baxter
- Section of Pediatric Hematology-Oncology, Texas Children's Hospital, Houston, TX, USA
| | - Suzanne Laughlin
- Department of Medical Imaging, The Hospital for Sick Children, Toronto, ON, Canada
| | - Zoltán Patay
- Department of Radiology, St Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Katherine E Warren
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
21
|
Kandels D, Pietsch T, Bison B, Warmuth-Metz M, Thomale UW, Kortmann RD, Timmermann B, Hernáiz Driever P, Witt O, Schmidt R, Gnekow AK. Loss of efficacy of subsequent nonsurgical therapy after primary treatment failure in pediatric low-grade glioma patients-Report from the German SIOP-LGG 2004 cohort. Int J Cancer 2020; 147:3471-3489. [PMID: 32580249 DOI: 10.1002/ijc.33170] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/02/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022]
Abstract
First-line treatment of pediatric low-grade glioma using surgery, radio- or chemotherapy fails in a relevant proportion of patients. We analyzed efficacy of subsequent surgical and nonsurgical therapies of the German cohort of the SIOP-LGG 2004 study (2004-2012, 1558 registered patients; median age at diagnosis 7.6 years, median observation time 9.2 years, overall survival 98%/96% at 5/10 years, 15% neurofibromatosis type 1 [NF1]). During follow-up, 1078/1558 patients remained observed without (n = 217), with 1 (n = 707), 2 (n = 124) or 3 to 6 (n = 30) tumor volume reductions; 480/1558 had 1 (n = 332), 2 (n = 80), 3 or more (n = 68) nonsurgical treatment-lines, accompanied by up to 4 tumor-reductive surgeries in 215/480; 265/480 patients never underwent any neurosurgical tumor volume reduction (163/265 optic pathway glioma). Patients with progressing tumors after first-line adjuvant treatment were at increased risk of suffering further progressions. Risk factors were young age (<1 year) at start of treatment, tumor dissemination or progression within 18 months after start of chemotherapy. Progression-free survival rates declined with subsequent treatment-lines, yet remaining higher for patients with NF1. In non-NF1-associated tumors, vinblastine monotherapy vs platinum-based chemotherapy was noticeably less effective when used as second-line treatment. Yet, for the entire cohort, results did not favor a certain sequence of specific treatment options. Rather, all can be aligned as a portfolio of choices which need careful balancing of risks and benefits. Future molecular data may predict long-term tumor biology.
Collapse
Affiliation(s)
- Daniela Kandels
- Swabian Children's Cancer Center, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Torsten Pietsch
- Institute of Neuropathology, DGNN Brain Tumor Reference Center, University of Bonn, Bonn, Germany
| | - Brigitte Bison
- Institute of Diagnostic and Interventional Neuroradiology, University Hospital Würzburg, Würzburg, Germany
| | - Monika Warmuth-Metz
- Institute of Diagnostic and Interventional Neuroradiology, University Hospital Würzburg, Würzburg, Germany
| | - Ulrich-Wilhelm Thomale
- Pediatric Neurosurgery, Charité Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | | | - Beate Timmermann
- Department of Particle Therapy, University Hospital Essen, West German Proton Therapy Centre Essen (WPE), West German Cancer Center (WTZ), German Cancer Consortium (DKTK), Essen, Germany
| | - Pablo Hernáiz Driever
- Department of Pediatric Oncology/Hematology, Charité Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Olaf Witt
- Hopp Children's Cancer Center Heidelberg (KiTZ), German Cancer Research Center (DKFZ), and Heidelberg University Hospital, Heidelberg, Germany
| | - René Schmidt
- Institute of Biostatistics and Clinical Research, University of Münster, Münster, Germany
| | - Astrid K Gnekow
- Swabian Children's Cancer Center, Medical Faculty, University of Augsburg, Augsburg, Germany
| |
Collapse
|
22
|
Abdalla G, Hammam A, Anjari M, D'Arco DF, Bisdas DS. Glioma surveillance imaging: current strategies, shortcomings, challenges and outlook. BJR Open 2020; 2:20200009. [PMID: 33178973 PMCID: PMC7594888 DOI: 10.1259/bjro.20200009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 01/11/2023] Open
Abstract
Inaccurate assessment of surveillance imaging to assess response to glioma therapy may have life-changing consequences. Varied management plans including chemotherapy, radiotherapy or immunotherapy may all contribute to heterogeneous post-treatment appearances and the overlap between the morphological features of pseudoprogression, pseudoresponse and radiation necrosis can make their discrimination very challenging. Therefore, there has been a drive to develop objective strategies for post-treatment assessment of brain gliomas. This review discusses the most important of these approaches such as the RANO "Response Assessment in Neuro-Oncology", iRANO "Immunotherapy Response Assessment in Neuro-Oncology" and RAPNO "Response Assessment in Paediatric Neuro-Oncology" models. In addition to these systematic approaches for glioma surveillance, the relatively limited information provided by conventional imaging modalities alone has motivated the development of novel advanced magnetic resonance (MR) and metabolic imaging methods for further discrimination between viable tumour and treatment induced changes. Multiple clinical trials and meta-analyses have investigated the diagnostic performance of these novel techniques in the follow up of brain gliomas, including both single modality descriptive studies and comparative imaging assessment. In this manuscript, we review the literature and discuss the promises and pitfalls of frequently studied modalities in glioma surveillance imaging, including MR perfusion, MR diffusion and MR spectroscopy. In addition, we evaluate other promising MR techniques such as chemical exchange saturation transfer as well as fludeoxyglucose and non-FDG positron emission tomography techniques.
Collapse
Affiliation(s)
- Gehad Abdalla
- Department of Neuroradiology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Trust, London, UK
| | - Ahmed Hammam
- Department of Neuroradiology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Trust, London, UK
| | - Mustafa Anjari
- Department of Neuroradiology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Trust, London, UK
| | - Dr. Felice D'Arco
- Department of Neuroradiology, Great Ormond Street Hospital for Children, London, UK
| | | |
Collapse
|
23
|
Nobre L, Zapotocky M, Ramaswamy V, Ryall S, Bennett J, Alderete D, Balaguer Guill J, Baroni L, Bartels U, Bavle A, Bornhorst M, Boue DR, Canete A, Chintagumpala M, Coven SL, Cruz O, Dahiya S, Dirks P, Dunkel IJ, Eisenstat D, Faure Conter C, Finch E, Finlay JL, Frappaz D, Garre ML, Gauvain K, Bechensteen AG, Hansford JR, Harting I, Hauser P, Hazrati LN, Huang A, Injac SG, Iurilli V, Karajannis M, Kaur G, Kyncl M, Krskova L, Laperriere N, Larouche V, Lassaletta A, Leary S, Lin F, Mascelli S, McKeown T, Milde T, Morales La Madrid A, Morana G, Morse H, Mushtaq N, Osorio DS, Packer R, Pavelka Z, Quiroga-Cantero E, Rutka J, Sabel M, Salgado D, Solano P, Sterba J, Su J, Sumerauer D, Taylor MD, Toledano H, Tsang DS, Valente Fernandes M, van Landeghem F, van Tilburg CM, Wilson B, Witt O, Zamecnik J, Bouffet E, Hawkins C, Tabori U. Outcomes of BRAF V600E Pediatric Gliomas Treated With Targeted BRAF Inhibition. JCO Precis Oncol 2020; 4:1900298. [PMID: 32923898 DOI: 10.1200/po.19.00298] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2020] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Children with pediatric gliomas harboring a BRAF V600E mutation have poor outcomes with current chemoradiotherapy strategies. Our aim was to study the role of targeted BRAF inhibition in these tumors. PATIENTS AND METHODS We collected clinical, imaging, molecular, and outcome information from patients with BRAF V600E-mutated glioma treated with BRAF inhibition across 29 centers from multiple countries. RESULTS Sixty-seven patients were treated with BRAF inhibition (pediatric low-grade gliomas [PLGGs], n = 56; pediatric high-grade gliomas [PHGGs], n = 11) for up to 5.6 years. Objective responses were observed in 80% of PLGGs, compared with 28% observed with conventional chemotherapy (P < .001). These responses were rapid (median, 4 months) and sustained in 86% of tumors up to 5 years while receiving therapy. After discontinuation of BRAF inhibition, 76.5% (13 of 17) of patients with PLGG experienced rapid progression (median, 2.3 months). However, upon rechallenge with BRAF inhibition, 90% achieved an objective response. Poor prognostic factors in conventional therapies, such as concomitant homozygous deletion of CDKN2A, were not associated with lack of response to BRAF inhibition. In contrast, only 36% of those with PHGG responded to BRAF inhibition, with all but one tumor progressing within 18 months. In PLGG, responses translated to 3-year progression-free survival of 49.6% (95% CI, 35.3% to 69.5%) versus 29.8% (95% CI, 20% to 44.4%) for BRAF inhibition versus chemotherapy, respectively (P = .02). CONCLUSION Use of BRAF inhibition results in robust and durable responses in BRAF V600E-mutated PLGG. Prospective studies are required to determine long-term survival and functional outcomes with BRAF inhibitor therapy in childhood gliomas.
Collapse
Affiliation(s)
- Liana Nobre
- Department of Hematology and Oncology, Hospital for Sick Children, Toronto, ON, Canada
| | - Michal Zapotocky
- Department of Hematology and Oncology, Hospital for Sick Children, Toronto, ON, Canada.,Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Vijay Ramaswamy
- Department of Hematology and Oncology, Hospital for Sick Children, Toronto, ON, Canada.,Developmental and Stem Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Scott Ryall
- Department of Hematology and Oncology, Hospital for Sick Children, Toronto, ON, Canada
| | - Julie Bennett
- Department of Hematology and Oncology, Hospital for Sick Children, Toronto, ON, Canada
| | - Daniel Alderete
- Hospital of Pediatrics SAMIC Prof. Dr Juan P. Garrahan, Buenos Aires, Argentina
| | - Julia Balaguer Guill
- Hospital Universitario y Politecnico La Fe, University of Valencia, Valencia, Spain
| | - Lorena Baroni
- Hospital of Pediatrics SAMIC Prof. Dr Juan P. Garrahan, Buenos Aires, Argentina
| | - Ute Bartels
- Department of Hematology and Oncology, Hospital for Sick Children, Toronto, ON, Canada
| | - Abhishek Bavle
- Jimmy Everest Section of Pediatric Heamatology/Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | | | - Daniel R Boue
- Nationwide Children's Hospital and Ohio State University, Columbus, OH
| | - Adela Canete
- Hospital Universitario y Politecnico La Fe, University of Valencia, Valencia, Spain
| | | | - Scott L Coven
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Indiana University, Indianapolis, IN
| | | | - Sonika Dahiya
- Washington University School of Medicine, St Louis, MO
| | - Peter Dirks
- Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, ON, Canada.,Division of Neurosurgery, Hospital for Sick Children, Toronto, ON, Canada
| | - Ira J Dunkel
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - David Eisenstat
- Stollery Children's Hospital, University of Alberta, Edmonton, AB, Canada
| | | | - Elizabeth Finch
- University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Jonathan L Finlay
- Nationwide Children's Hospital and Ohio State University, Columbus, OH
| | - Didier Frappaz
- Institute d'Hémato-Oncologie Pédiatrique, Centre Leon Berard, Lyon, France
| | | | - Karen Gauvain
- Washington University School of Medicine, St Louis, MO
| | | | - Jordan R Hansford
- Royal Children's Hospital, Murdoch Children's Research Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Inga Harting
- Hopp Children's Cancer Center Heidelberg, Heidelberg, Germany
| | | | - Lili-Naz Hazrati
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Annie Huang
- Department of Hematology and Oncology, Hospital for Sick Children, Toronto, ON, Canada.,Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, ON, Canada
| | | | | | | | | | - Martin Kyncl
- Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Lenka Krskova
- Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Normand Laperriere
- Department of Hematology and Oncology, Hospital for Sick Children, Toronto, ON, Canada
| | | | | | | | - Frank Lin
- Texas Children's Cancer Center, Houston, TX
| | | | - Tara McKeown
- Department of Hematology and Oncology, Hospital for Sick Children, Toronto, ON, Canada
| | - Till Milde
- Hopp Children's Cancer Center Heidelberg, Heidelberg, Germany
| | | | | | | | | | - Diana S Osorio
- Nationwide Children's Hospital and Ohio State University, Columbus, OH
| | - Roger Packer
- Children's National Health System, Washington, DC
| | - Zdenek Pavelka
- University Hospital Brno, Masaryk University, and ICRC Brno, Brno, Czech Republic
| | | | - James Rutka
- Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, ON, Canada.,Division of Neurosurgery, Hospital for Sick Children, Toronto, ON, Canada
| | - Magnus Sabel
- Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | | | - Palma Solano
- Hospital Infantil Virgen del Rocío, Sevilla, Spain
| | - Jaroslav Sterba
- University Hospital Brno, Masaryk University, and ICRC Brno, Brno, Czech Republic
| | - Jack Su
- Texas Children's Cancer Center, Houston, TX
| | - David Sumerauer
- Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Michael D Taylor
- Developmental and Stem Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada.,Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, ON, Canada.,Division of Neurosurgery, Hospital for Sick Children, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Department of Surgery, University of Ontario, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Helen Toledano
- Schneiders Children's Medical Center of Israel, Petah Tikva, Israel
| | - Derek S Tsang
- Department of Hematology and Oncology, Hospital for Sick Children, Toronto, ON, Canada
| | | | | | | | - Bev Wilson
- Stollery Children's Hospital, University of Alberta, Edmonton, AB, Canada
| | - Olaf Witt
- Hopp Children's Cancer Center Heidelberg, Heidelberg, Germany
| | - Josef Zamecnik
- Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Eric Bouffet
- Department of Hematology and Oncology, Hospital for Sick Children, Toronto, ON, Canada
| | - Cynthia Hawkins
- Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Department of Pediatric Laboratory Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Uri Tabori
- Department of Hematology and Oncology, Hospital for Sick Children, Toronto, ON, Canada.,Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
24
|
Gilligan LA, DeWire-Schottmiller MD, Fouladi M, DeBlank P, Leach JL. Tumor Response Assessment in Diffuse Intrinsic Pontine Glioma: Comparison of Semiautomated Volumetric, Semiautomated Linear, and Manual Linear Tumor Measurement Strategies. AJNR Am J Neuroradiol 2020; 41:866-873. [PMID: 32354716 DOI: 10.3174/ajnr.a6555] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/26/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND PURPOSE 2D measurements of diffuse intrinsic pontine gliomas are limited by variability, and volumetric response criteria are poorly defined. Semiautomated 2D measurements may improve consistency; however, the impact on tumor response assessments is unknown. The purpose of this study was to compare manual 2D, semiautomated 2D, and volumetric measurement strategies for diffuse intrinsic pontine gliomas. MATERIALS AND METHODS This study evaluated patients with diffuse intrinsic pontine gliomas through a Phase I/II trial (NCT02607124). Clinical 2D cross-product values were derived from manual linear measurements (cross-product = long axis × short axis). By means of dedicated software (mint Lesion), tumor margins were traced and maximum cross-product and tumor volume were automatically derived. Correlation and bias between methods were assessed, and response assessment per measurement strategy was reported. RESULTS Ten patients (median age, 7.6 years) underwent 58 MR imaging examinations. Correlation and mean bias (95% limits) of percentage change in tumor size from prior examinations were the following: clinical and semiautomated cross-product, r = 0.36, -1.5% (-59.9%, 56.8%); clinical cross-product and volume, r = 0.61, -2.1% (-52.0%, 47.8%); and semiautomated cross-product and volume, r = 0.79, 0.6% (-39.3%, 38.1%). Stable disease, progressive disease, and partial response rates per measurement strategy were the following: clinical cross-product, 82%, 18%, 0%; semiautomated cross-product, 54%, 42%, 4%; and volume, 50%, 46%, 4%, respectively. CONCLUSIONS Manual 2D cross-product measurements may underestimate tumor size and disease progression compared with semiautomated 2D and volumetric measurements.
Collapse
Affiliation(s)
- L A Gilligan
- From the Departments of Radiology (L.A.G., J.L.L.).,Department of Graduate Medical Education (L.A.G., M.D.D.-S.), Mount Carmel Health System, Columbus, Ohio
| | - M D DeWire-Schottmiller
- and Cancer and Blood Diseases Institute (M.D.D.-S., M.F.), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Graduate Medical Education (L.A.G., M.D.D.-S.), Mount Carmel Health System, Columbus, Ohio
| | - M Fouladi
- and Cancer and Blood Diseases Institute (M.D.D.-S., M.F.), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,and Departments of Pediatrics (M.F., P.D.)
| | - P DeBlank
- and Departments of Pediatrics (M.F., P.D.)
| | - J L Leach
- From the Departments of Radiology (L.A.G., J.L.L.) .,Radiology (J.L.L.), University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
25
|
Dietz MS, Beach CZ, Barajas R, Parappilly MS, Sengupta SK, Baird LC, Ciporen JN, Han SJ, Loret de Mola R, Cho YJ, Nazemi KJ, McClelland S, Wong MH, Jaboin JJ. Measure Twice: Promise of Liquid Biopsy in Pediatric High-Grade Gliomas. Adv Radiat Oncol 2020; 5:152-162. [PMID: 32280814 PMCID: PMC7136635 DOI: 10.1016/j.adro.2019.12.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/07/2019] [Accepted: 12/17/2019] [Indexed: 12/20/2022] Open
Abstract
Purpose To review and critique the current state of liquid biopsy in pHGG. Materials and Methods Published literature was reviewed for articles related to liquid biopsy in pediatric glioma and adult glioma with a focus on high-grade gliomas. Results This review discusses the current state of liquid biomarkers of pHGG and their potential applications for liquid biopsy development. Conclusions While nascent, the progress toward identifying circulating analytes of pHGG primes the field of neuro-oncoogy for liquid biopsy development.
Collapse
Affiliation(s)
- Matthew S Dietz
- Department of Pediatrics, Oregon Health & Science University, Portland, Oregon
| | - Catherine Z Beach
- Department of Radiation Medicine, Oregon Health & Science University, Portland, Oregon
| | - Ramon Barajas
- Department of Diagnostic Radiology, Oregon Health & Science University, Portland, Oregon.,Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon
| | - Michael S Parappilly
- Department of Diagnostic Radiology, Oregon Health & Science University, Portland, Oregon
| | - Sidharth K Sengupta
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Lissa C Baird
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon
| | - Jeremy N Ciporen
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon
| | - Seunggu J Han
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon
| | | | - Yoon Jae Cho
- Department of Neurology, Oregon Health & Science University, Portland, Oregon.,The Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Kellie J Nazemi
- Department of Pediatrics, Oregon Health & Science University, Portland, Oregon
| | - Shearwood McClelland
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Melissa H Wong
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon.,The Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Jerry J Jaboin
- Department of Radiation Medicine, Oregon Health & Science University, Portland, Oregon.,The Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
26
|
Trunin YY, Golanov AV, Konovalov AN, Pronin IN. [Pseudoprogression of intracranial pilocytic astrocytomas and other low-grade gliomas. Literature review and case report]. ZHURNAL VOPROSY NEIROKHIRURGII IMENI N. N. BURDENKO 2020; 84:105-111. [PMID: 33306307 DOI: 10.17116/neiro202084061105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Tumor pseudoprogression is characterized by temporary tumor enlargement following radiotherapy with subsequent stabilization or regression without additional treatment. This phenomenon has been comprehensively described in patients with malignant gliomas. However, this phenomenon has not been sufficiently studied in patients with low-grade gliomas including pilocytic astrocytomas. In recent years, more and more researches devoted to this problem have appeared in the literature. It seems relevant to conduct a meta-analysis of these data in the modern literature.
Collapse
Affiliation(s)
| | - A V Golanov
- Burdenko Neurosurgical Center, Moscow, Russia
| | | | - I N Pronin
- Burdenko Neurosurgical Center, Moscow, Russia
| |
Collapse
|
27
|
|
28
|
Calmon R, Puget S, Varlet P, Dangouloff-Ros V, Blauwblomme T, Beccaria K, Grevent D, Sainte-Rose C, Castel D, Debily MA, Dufour C, Bolle S, Dhermain F, Saitovitch A, Zilbovicius M, Brunelle F, Grill J, Boddaert N. Cerebral blood flow changes after radiation therapy identifies pseudoprogression in diffuse intrinsic pontine gliomas. Neuro Oncol 2019; 20:994-1002. [PMID: 29244086 DOI: 10.1093/neuonc/nox227] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background The interval between progression and death in diffuse intrinsic pontine glioma (DIPG) is usually <6 months. However, reports of longer patient survival following radiotherapy, in the presence of radiological signs of progression, suggest that these cases may be comparable to pseudoprogression observed in adult glioblastoma. Our aim was to identify such cases and compare their multimodal MRI features with those of patients who did not present the same evolution. Methods Multimodal MRIs of 43 children treated for DIPG were retrospectively selected at 4 timepoints: baseline, after radiotherapy, during true progression, and at the last visit. The patients were divided into 2 groups depending on whether they presented conventional MRI changes that mimicked progression. The apparent diffusion coefficient, arterial spin labeling cerebral blood flow (ASL-CBF), and dynamic susceptibility contrast perfusion relative cerebral blood volume (DSCrCBV) and flow (DSCrCBF) values were recorded for each tumor voxel, avoiding necrotic areas. Results After radiotherapy, 19 patients (44%) showed radiological signs that mimicked progression: 16 survived >6 months following so-called pseudoprogression, with a median of 8.9 months and a maximum of 35.6 months. All 43 patients exhibited increased blood volume and flow after radiotherapy, but the 90th percentile of those with signs of pseudoprogression had a greater increase of ASL-CBF (P < 0.001). Survival between the 2 groups did not differ significantly. During true progression, DSCrCBF and DSCrCBV values increased only in patients who had not experienced pseudoprogression. Conclusions Pseudoprogression is a frequent phenomenon in DIPG patients. This condition needs to be recognized before considering treatment discontinuation. In this study, the larger increase of the ASL-CBF ratio after radiotherapy accurately distinguished pseudoprogression from true progression.
Collapse
Affiliation(s)
- Raphael Calmon
- Hôpital Necker Enfants Malades, Pediatric Radiology Department, Paris, France.,Imagine: Institut de Maladies Génétiques, Paris, France.,INSERM, Paris, France.,Université Paris Descartes, ComUE Sorbonne Paris Cité, Paris, France
| | - Stephanie Puget
- Hôpital Necker Enfants Malades, Pediatric Neurosurgery Department, Paris, France
| | - Pascale Varlet
- INSERM, Paris, France.,Centre Hospitalier Sainte-Anne, Laboratoire de Neuropathologie, Paris, France
| | - Volodia Dangouloff-Ros
- Hôpital Necker Enfants Malades, Pediatric Radiology Department, Paris, France.,Imagine: Institut de Maladies Génétiques, Paris, France.,INSERM, Paris, France.,Université Paris Descartes, ComUE Sorbonne Paris Cité, Paris, France
| | - Thomas Blauwblomme
- Hôpital Necker Enfants Malades, Pediatric Neurosurgery Department, Paris, France
| | - Kevin Beccaria
- Hôpital Necker Enfants Malades, Pediatric Neurosurgery Department, Paris, France
| | - David Grevent
- Hôpital Necker Enfants Malades, Pediatric Radiology Department, Paris, France.,Imagine: Institut de Maladies Génétiques, Paris, France.,INSERM, Paris, France.,Université Paris Descartes, ComUE Sorbonne Paris Cité, Paris, France
| | | | - David Castel
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8203 et Universite Paris Saclay, Villejuif, France
| | - Marie-Anne Debily
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8203 et Universite Paris Saclay, Villejuif, France.,Université Evry Val-d'Essonne, Département de Biologie, Evry, France
| | - Christelle Dufour
- Gustave Roussy, Département de Cancerologie de l'Enfant et de l'Adolescent, Villejuif, France
| | - Stéphanie Bolle
- Gustave Roussy, Département de Radiothérapie, Villejuif, France
| | - Frederic Dhermain
- Gustave Roussy, Département de Cancerologie de l'Enfant et de l'Adolescent, Villejuif, France.,Gustave Roussy, Département de Radiothérapie, Villejuif, France
| | - Ana Saitovitch
- Imagine: Institut de Maladies Génétiques, Paris, France.,INSERM, Paris, France
| | | | - Francis Brunelle
- Hôpital Necker Enfants Malades, Pediatric Radiology Department, Paris, France.,Imagine: Institut de Maladies Génétiques, Paris, France.,INSERM, Paris, France.,Université Paris Descartes, ComUE Sorbonne Paris Cité, Paris, France
| | - Jacques Grill
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8203 et Universite Paris Saclay, Villejuif, France.,Gustave Roussy, Département de Cancerologie de l'Enfant et de l'Adolescent, Villejuif, France
| | - Nathalie Boddaert
- Hôpital Necker Enfants Malades, Pediatric Radiology Department, Paris, France.,Imagine: Institut de Maladies Génétiques, Paris, France.,INSERM, Paris, France.,Université Paris Descartes, ComUE Sorbonne Paris Cité, Paris, France
| |
Collapse
|
29
|
Bevacizumab-containing regimen in relapsed/progressed brain tumors: a single-institution experience. Childs Nerv Syst 2019; 35:1007-1012. [PMID: 30903281 DOI: 10.1007/s00381-019-04117-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/08/2019] [Indexed: 01/01/2023]
Abstract
AIM The aim of the study is to assess tumor response, treatment-related toxicities, progression-free survival (PFS), and overall survival (OS) in patients with relapsed/refractory brain tumors treated with bevacizumab-containing regimen. METHODS Patients that had received I and II line treatments with or without megatherapy were included. Doses and schedule were as follows: bevacizumab (BVZ) 10 mg/kg i.v. with irinotecan (IRI) 150 mg/m2 i.v. every 2 weeks ± temozolamide (TMZ) 200 mg/m2 p.o. daily for 5 days every 4 weeks. TMZ was omitted in heavily pretreated cases. RESULTS Between 2013 and 2018, 12 patients (3F/9M), median age 161 months (range 66-348), affected with medulloblastoma (n 7), or low-grade glioma (n 2), or high-grade glioma (n 3), received BVZ/IRI association (median courses 20, range 4-67); 3 of them continued single-agent BVZ (median courses 23, range 8-39). TMZ (median courses 8, range 2-26) was administered in eight patients and then stopped in three of them because of myelotoxicity or lack of compliance. Treatment was well tolerated. After 3 months, two complete responses, two partial responses, seven stable diseases, and one progressive disease were observed. Nine cases experienced an improvement in neurological symptoms. Median time to progression was 11 months (95% confidence interval, 4-18 months). Six-month and 2-year PFS were 75% and 42%, respectively. The OS is 33%; interestingly, two cases (one medulloblastoma and one high-grade glioma) are progression-free off-therapy since 30 and 48 months, respectively. CONCLUSIONS BVZ/IRI association ± TMZ showed encouraging therapeutic activity and low toxicity in this series of relapsed/refractory brain tumors.
Collapse
|
30
|
Bouzidi Y, Barteau E, Lejeune J, Dejobert M, Gravellier B, Sirinelli D, Cottier JP, Morel B. Detection of recurrent brain tumors in children: No significant difference in sensitivity between unenhanced and contrast-enhanced MRI. Neuroradiol J 2019; 32:259-266. [PMID: 31017042 DOI: 10.1177/1971400919845619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Magnetic resonance imaging (MRI) with a gadolinium injection is currently used in the follow-up of children in remission of cerebral tumors (CTs). Intracerebral gadolinium deposition has been recently reported with unknown risks. The aim of this study was to evaluate the sensitivity of unenhanced brain MRI (U-MRI) in detection of tumor recurrence. METHODS AND MATERIALS A set of 58 U-MRIs of children in remission was retrospectively evaluated by three seniors (a neuroradiologist, a pediatric and a general radiologist) and one junior to look for any recurrence. Clinical, tumoral and imaging data were collected. The final diagnosis was anatomopathological when available, or the clinicoradiological evolution. Sensitivity, specificity, predictive values and interobserver agreement were calculated. A Fisher test and Fleiss kappa coefficient were performed. RESULTS For the seniors, the U-MRI had a sensitivity of 81% (95% confidence interval (CI): 0.56-0.90), and a negative predictive value (NPV) of 82% (95% CI: 0.63-0.94). The U-MRI sensitivity, regardless of the observer, was not significantly different from the contrast-enhanced MRI sensitivity (86%) according to a Fisher test (p > 0.05). No significant difference in sensitivity within the subgroups was found. The interobserver agreement of seniors was good (κ = 0.68). CONCLUSION U-MRI brain was suboptimal for 80% of patients. Three-dimensional millimetric, fluid-attenuated inversion recovery, and diffusion would constitute helpful sequences in follow-up. Further specific studies depending on each tumor type are still required to determine whether a potential abstention of gadolinium intravenous injection should be discussed for children.
Collapse
Affiliation(s)
- Yasmina Bouzidi
- 1 Pediatric Radiology Department, Clocheville Hospital, University Hospital Center of Tours, France
| | - Emmanuel Barteau
- 1 Pediatric Radiology Department, Clocheville Hospital, University Hospital Center of Tours, France
| | - Julien Lejeune
- 2 Pediatric Oncologic Department, Clocheville Hospital, University Hospital Center of Tours, France
| | - Maelle Dejobert
- 3 Neuroradiology Department, Bretonneau Hospital, University Hospital Center of Tours, France
| | - Bastien Gravellier
- 1 Pediatric Radiology Department, Clocheville Hospital, University Hospital Center of Tours, France
| | - Dominique Sirinelli
- 1 Pediatric Radiology Department, Clocheville Hospital, University Hospital Center of Tours, France.,4 Faculty of Medicine, Francois Rabelais University, Tours, France
| | - Jean Philippe Cottier
- 3 Neuroradiology Department, Bretonneau Hospital, University Hospital Center of Tours, France.,4 Faculty of Medicine, Francois Rabelais University, Tours, France
| | - Baptiste Morel
- 1 Pediatric Radiology Department, Clocheville Hospital, University Hospital Center of Tours, France.,4 Faculty of Medicine, Francois Rabelais University, Tours, France
| |
Collapse
|
31
|
Colafati GS, Voicu IP, Carducci C, Caulo M, Vinci M, Diomedi-Camassei F, Merli P, Carai A, Miele E, Cacchione A, Tomà P, Locatelli F, Mastronuzzi A. Direct Involvement of Cranial Nerve V at Diagnosis in Patients With Diffuse Intrinsic Pontine Glioma: A Potential Magnetic Resonance Predictor of Short-Term Survival. Front Oncol 2019; 9:204. [PMID: 31019890 PMCID: PMC6458256 DOI: 10.3389/fonc.2019.00204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 03/11/2019] [Indexed: 12/05/2022] Open
Abstract
Background: Diffuse intrinsic pontine glioma (DIPG) has a dismal prognosis. Magnetic resonance imaging (MRI) remains the gold standard for non-invasive DIPG diagnosis. MRI features have been tested as surrogate biomarkers. We investigated the direct involvement of cranial nerve V (CN V) in DIPG at diagnosis and its utility as predictor of poor overall survival. Materials and Methods: We examined MRI scans of 35 consecutive patients with radiological diagnosis of DIPG. Direct involvement of CN V was assessed on the diagnostic scans. Differences in overall survival (OS) and time to progression (TTP) were analyzed for involvement of CN V, sex, age, tumor size, ring enhancement, and treatment regimen. Correlations between involvement of CN V and disease dissemination, magnet strength and slice thickness were analyzed. Statistical analyses included Kaplan-Meier curves, log-rank test and Spearman's Rho. Results: After excluding six long-term survivors, 29 patients were examined (15 M, 14 F). Four patients presented direct involvement of CN V. Histological data were available in 12 patients. Median OS was 11 months (range 3-23 months). Significant differences in OS were found for direct involvement of CN V (median OS: 7 months, 95% CI 1.1-12.9 months for involvement of CN V vs. 13 months, 95% CI 10.2-15.7 for lack of involvement of CN V, respectively, p < 0.049). Significant differences in TTP were found for the two treatment regimens (median TTP: 4 months, 95% CI 2.6-5.3 vs. 7 months, 95% CI 5.9-8.1, respectively, p < 0.027). No significant correlation was found between involvement of CN V and magnet strength or slice thickness (r = -0.201; p = NS). A trend toward positive correlation was found between direct involvement of CN V at diagnosis and dissemination of disease at follow-up (r = 0.347; p < 0.065). Conclusions: In our cohort, direct involvement of CN V correlated with poor prognosis. Based on our data, we suggest that in DIPG direct involvement of CN V should be routinely evaluated on diagnostic scans.
Collapse
Affiliation(s)
| | - Ioan Paul Voicu
- Department of Imaging, Neuroradiology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Chiara Carducci
- Department of Imaging, Neuroradiology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Massimo Caulo
- Department of Neuroscience and Imaging, ITAB-Institute of Advanced Biomedical Technologies, University G. d'Annunzio, Chieti, Italy
| | - Maria Vinci
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Pietro Merli
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Andrea Carai
- Neurosurgery Unit, Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Evelina Miele
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Antonella Cacchione
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Paolo Tomà
- Radiology Unit, Department of Imaging, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Franco Locatelli
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Angela Mastronuzzi
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
32
|
|
33
|
Chukwueke UN, Wen PY. Use of the Response Assessment in Neuro-Oncology (RANO) criteria in clinical trials and clinical practice. CNS Oncol 2019; 8:CNS28. [PMID: 30806082 PMCID: PMC6499019 DOI: 10.2217/cns-2018-0007] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/15/2018] [Indexed: 11/21/2022] Open
Affiliation(s)
- Ugonma N Chukwueke
- Department of Medical Oncology, Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Neurology, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Patrick Y Wen
- Department of Medical Oncology, Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Neurology, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
| |
Collapse
|
34
|
Warren KE, Vezina G, Poussaint TY, Warmuth-Metz M, Chamberlain MC, Packer RJ, Brandes AA, Reiss M, Goldman S, Fisher MJ, Pollack IF, Prados MD, Wen PY, Chang SM, Dufour C, Zurakowski D, Kortmann RD, Kieran MW. Response assessment in medulloblastoma and leptomeningeal seeding tumors: recommendations from the Response Assessment in Pediatric Neuro-Oncology committee. Neuro Oncol 2019; 20:13-23. [PMID: 28449033 DOI: 10.1093/neuonc/nox087] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Lack of standard response criteria in clinical trials for medulloblastoma and other seeding tumors complicates assessment of therapeutic efficacy and comparisons across studies. An international working group was established to develop consensus recommendations for response assessment. The aim is that these recommendations be prospectively evaluated in clinical trials, with the goal of achieving more reliable risk stratification and uniformity across clinical trials. Current practices and literature review were performed to identify major confounding issues and justify subsequently developed recommendations; in areas lacking scientific investigations, recommendations were based on experience of committee members and consensus was reached after discussion. Recommendations apply to both adult and pediatric patients with medulloblastoma and other seeding tumors. Response should be assessed using MR imaging (brain and spine), CSF cytology, and neurologic examination. Clinical imaging standards with minimum mandatory sequence acquisition that optimizes detection of leptomeningeal metastases are defined. We recommend central review prior to inclusion in treatment cohorts to ensure appropriate risk stratification and cohort inclusion. Consensus recommendations and response definitions for patients with medulloblastomas and other seeding tumors have been established; as with other Response Assessment in Neuro-Oncology recommendations, these need to now be prospectively validated in clinical trials.
Collapse
Affiliation(s)
- Katherine E Warren
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | - Gilbert Vezina
- Department of Radiology, Children's National Medical Center, Washington, DC
| | - Tina Y Poussaint
- Department of Radiology, Boston Children's Hospital, Boston, Massachusetts
| | - Monika Warmuth-Metz
- Department of Neuroradiology, University Hospital Würzburg, Würzburg, Germany
| | - Marc C Chamberlain
- Department of Neurology, Seattle Cancer Care Alliance, Seattle, Washington
| | - Roger J Packer
- Center for Neuroscience and Behavioral Medicine, Children's National Medical Center, Washington, DC
| | - Alba A Brandes
- Medical Oncology Department, AUSL-IRCCS Scienze Neurologiche, Bologna, Italy
| | - Moshe Reiss
- Division of Pediatric Neuro-Oncology, New York Medical College, Valhalla, New York
| | - Stewart Goldman
- Hematology-Oncology, Neuro-Oncology & Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Michael J Fisher
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Ian F Pollack
- Department of Neurological Surgery, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Michael D Prados
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California.,Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Susan M Chang
- Department of Neurosurgery, University of California San Francisco, San Francisco, California
| | - Christelle Dufour
- Department of Pediatric and Adolescent Oncology, Gustave Roussy, Villejuif, France
| | - David Zurakowski
- Departments of Anesthesia & Surgery, Boston Children's Hospital, Boston, Massachusetts
| | - Rolf D Kortmann
- Department of Radiation Oncology, University of Leipzig, Leipzig, Germany
| | - Mark W Kieran
- Pediatric Neuro-Oncology, Dana Farber Boston Children's Cancer and Blood Disorder's Center, Boston, Massachusetts
| |
Collapse
|
35
|
D'Arco F, Culleton S, De Cocker LJL, Mankad K, Davila J, Tamrazi B. Current concepts in radiologic assessment of pediatric brain tumors during treatment, part 1. Pediatr Radiol 2018; 48:1833-1843. [PMID: 29980859 DOI: 10.1007/s00247-018-4194-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 05/26/2018] [Accepted: 06/21/2018] [Indexed: 12/26/2022]
Abstract
Pediatric brain tumors differ from those in adults by location, phenotype and genotype. In addition, they show dissimilar imaging characteristics before and after treatment. While adult brain tumor treatment effects are primarily assessed on MRI by measuring the contrast-enhancing components in addition to abnormalities on T2-weighted and fluid-attenuated inversion recovery images, these methods cannot be simply extrapolated to pediatric central nervous system tumors. A number of researchers have attempted to solve the problem of tumor assessment during treatment in pediatric neuro-oncology; specifically, the Response Assessment in Pediatric Neuro-Oncology (RAPNO) working group was recently established to deal with the distinct challenges in evaluating treatment-related changes on imaging, but no established criteria are available. In this article we review the current methods to evaluate brain tumor therapy and the numerous challenges that remain. In part 1, we examine the role of T2-weighted imaging and fluid-attenuated inversion recovery sequences, contrast enhancement, volumetrics and diffusion imaging techniques. We pay particular attention to several specific pediatric brain tumors, such as optic pathway glioma, diffuse midline glioma and medulloblastoma. Finally, we review the best means to assess leptomeningeal seeding.
Collapse
Affiliation(s)
- Felice D'Arco
- Department of Neuroradiology, Great Ormond Street Hospital for Children NHS Trust, Great Ormond Street, London, WC1N 3JH, UK. felice.d'
| | - Sinead Culleton
- Department of Neuroradiology, Great Ormond Street Hospital for Children NHS Trust, Great Ormond Street, London, WC1N 3JH, UK
| | | | - Kshitij Mankad
- Department of Neuroradiology, Great Ormond Street Hospital for Children NHS Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Jorge Davila
- Department of Medical Imaging, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Benita Tamrazi
- Department of Radiology, Children's Hospital Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
36
|
Thust SC, Heiland S, Falini A, Jäger HR, Waldman AD, Sundgren PC, Godi C, Katsaros VK, Ramos A, Bargallo N, Vernooij MW, Yousry T, Bendszus M, Smits M. Glioma imaging in Europe: A survey of 220 centres and recommendations for best clinical practice. Eur Radiol 2018. [PMID: 29536240 PMCID: PMC6028837 DOI: 10.1007/s00330-018-5314-5] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Objectives At a European Society of Neuroradiology (ESNR) Annual Meeting 2015 workshop, commonalities in practice, current controversies and technical hurdles in glioma MRI were discussed. We aimed to formulate guidance on MRI of glioma and determine its feasibility, by seeking information on glioma imaging practices from the European Neuroradiology community. Methods Invitations to a structured survey were emailed to ESNR members (n=1,662) and associates (n=6,400), European national radiologists’ societies and distributed via social media. Results Responses were received from 220 institutions (59% academic). Conventional imaging protocols generally include T2w, T2-FLAIR, DWI, and pre- and post-contrast T1w. Perfusion MRI is used widely (85.5%), while spectroscopy seems reserved for specific indications. Reasons for omitting advanced imaging modalities include lack of facility/software, time constraints and no requests. Early postoperative MRI is routinely carried out by 74% within 24–72 h, but only 17% report a percent measure of resection. For follow-up, most sites (60%) issue qualitative reports, while 27% report an assessment according to the RANO criteria. A minority of sites use a reporting template (23%). Conclusion Clinical best practice recommendations for glioma imaging assessment are proposed and the current role of advanced MRI modalities in routine use is addressed. Key Points • We recommend the EORTC-NBTS protocol as the clinical standard glioma protocol. • Perfusion MRI is recommended for diagnosis and follow-up of glioma. • Use of advanced imaging could be promoted with increased education activities. • Most response assessment is currently performed qualitatively. • Reporting templates are not widely used, and could facilitate standardisation. Electronic supplementary material The online version of this article (10.1007/s00330-018-5314-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- S C Thust
- Lysholm Neuroradiology Department, National Hospital for Neurology and Neurosurgery, London, UK
- Department of Brain Rehabilitation and Repair, UCL Institute of Neurology, London, UK
- Imaging Department, University College London Hospital, London, UK
| | - S Heiland
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
| | - A Falini
- Department of Neuroradiology, San Raffaele Scientific Institute, Milan, Italy
| | - H R Jäger
- Lysholm Neuroradiology Department, National Hospital for Neurology and Neurosurgery, London, UK
- Department of Brain Rehabilitation and Repair, UCL Institute of Neurology, London, UK
- Imaging Department, University College London Hospital, London, UK
| | - A D Waldman
- Neuroimaging Sciences, University of Edinburgh, Edinburgh, UK
| | - P C Sundgren
- Institution for Clinical Sciences/Radiology, Lund University, Lund, Sweden
- Centre for Imaging and Physiology, Skåne University hospital, Lund, Sweden
| | - C Godi
- Department of Neuroradiology, San Raffaele Scientific Institute, Milan, Italy
| | - V K Katsaros
- General Anti-Cancer and Oncological Hospital "Agios Savvas", Athens, Greece
- Central Clinic of Athens, Athens, Greece
- University of Athens, Athens, Greece
| | - A Ramos
- Hospital 12 de Octubre, Madrid, Spain
| | - N Bargallo
- Image Diagnostic Centre, Hospital Clinic de Barcelona, Barcelona, Spain
- Magnetic Resonance Core Facility, Institut per la Recerca Biomedica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - M W Vernooij
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - T Yousry
- Lysholm Neuroradiology Department, National Hospital for Neurology and Neurosurgery, London, UK
| | - M Bendszus
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
| | - M Smits
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
37
|
D’Arco F, O’Hare P, Dashti F, Lassaletta A, Loka T, Tabori U, Talenti G, Thust S, Messalli G, Hales P, Bouffet E, Laughlin S. Volumetric assessment of tumor size changes in pediatric low-grade gliomas: feasibility and comparison with linear measurements. Neuroradiology 2018; 60:427-436. [DOI: 10.1007/s00234-018-1979-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 01/09/2018] [Indexed: 11/25/2022]
|
38
|
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
|
39
|
Lucas JT, Cooper DA, Hwang S, Tinkle C, Li X, Li Y, Orr B, Merchant TE, Broniscer A. Prognostic Relevance of Treatment Failure Patterns in Pediatric High-Grade Glioma: Is There a Role for a Revised Failure Classification System? Int J Radiat Oncol Biol Phys 2017; 99:450-458. [DOI: 10.1016/j.ijrobp.2017.04.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 02/24/2017] [Accepted: 04/26/2017] [Indexed: 10/19/2022]
|
40
|
Boult JKR, Box G, Vinci M, Perryman L, Eccles SA, Jones C, Robinson SP. Evaluation of the Response of Intracranial Xenografts to VEGF Signaling Inhibition Using Multiparametric MRI. Neoplasia 2017; 19:684-694. [PMID: 28780387 PMCID: PMC5547238 DOI: 10.1016/j.neo.2017.05.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/15/2017] [Accepted: 05/15/2017] [Indexed: 11/29/2022] Open
Abstract
Vascular endothelial growth factor A (VEGF-A) is considered one of the most important factors in tumor angiogenesis, and consequently, a number of therapeutics have been developed to inhibit VEGF signaling. Therapeutic strategies to target brain malignancies, both primary brain tumors, particularly in pediatric patients, and metastases, are lacking, but targeting angiogenesis may be a promising approach. Multiparametric MRI was used to investigate the response of orthotopic SF188luc pediatric glioblastoma xenografts to small molecule pan-VEGFR inhibitor cediranib and the effects of both cediranib and cross-reactive human/mouse anti-VEGF-A antibody B20-4.1.1 in intracranial MDA-MB-231 LM2–4 breast cancer xenografts over 48 hours. All therapeutic regimens resulted in significant tumor growth delay. In cediranib-treated SF188luc tumors, this was associated with lower Ktrans (compound biomarker of perfusion and vascular permeability) than in vehicle-treated controls. Cediranib also induced significant reductions in both Ktrans and apparent diffusion coefficient (ADC) in MDA-MB-231 LM2–4 tumors associated with decreased histologically assessed perfusion. B20-4.1.1 treatment resulted in decreased Ktrans, but in the absence of a change in perfusion; a non-significant reduction in vascular permeability, assessed by Evans blue extravasation, was observed in treated tumors. The imaging responses of intracranial MDA-MB-231 LM2–4 tumors to VEGF/VEGFR pathway inhibitors with differing mechanisms of action are subtly different. We show that VEGF pathway blockade resulted in tumor growth retardation and inhibition of tumor vasculature in preclinical models of pediatric glioblastoma and breast cancer brain metastases, suggesting that multiparametric MRI can provide a powerful adjunct to accelerate the development of antiangiogenic therapies for use in these patient populations.
Collapse
Affiliation(s)
- Jessica K R Boult
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, SM2 5NG, UK.
| | - Gary Box
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, SM2 5NG, UK.
| | - Maria Vinci
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, SM2 5NG, UK; Division of Molecular Pathology, The Institute of Cancer Research, London, SM2 5NG, UK.
| | - Lara Perryman
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, SM2 5NG, UK; Division of Molecular Pathology, The Institute of Cancer Research, London, SM2 5NG, UK.
| | - Suzanne A Eccles
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, SM2 5NG, UK.
| | - Chris Jones
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, SM2 5NG, UK; Division of Molecular Pathology, The Institute of Cancer Research, London, SM2 5NG, UK.
| | - Simon P Robinson
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, SM2 5NG, UK.
| |
Collapse
|
41
|
Wen PY, Chang SM, Van den Bent MJ, Vogelbaum MA, Macdonald DR, Lee EQ. Response Assessment in Neuro-Oncology Clinical Trials. J Clin Oncol 2017; 35:2439-2449. [PMID: 28640707 PMCID: PMC5516482 DOI: 10.1200/jco.2017.72.7511] [Citation(s) in RCA: 314] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Development of novel therapies for CNS tumors requires reliable assessment of response and progression. This requirement has been particularly challenging in neuro-oncology for which contrast enhancement serves as an imperfect surrogate for tumor volume and is influenced by agents that affect vascular permeability, such as antiangiogenic therapies. In addition, most tumors have a nonenhancing component that can be difficult to accurately quantify. To improve the response assessment in neuro-oncology and to standardize the criteria that are used for different CNS tumors, the Response Assessment in Neuro-Oncology (RANO) working group was established. This multidisciplinary international working group consists of neuro-oncologists, medical oncologists, neuroradiologists, neurosurgeons, radiation oncologists, neuropsychologists, and experts in clinical outcomes assessments, working in collaboration with government and industry to enhance the interpretation of clinical trials. The RANO working group was originally created to update response criteria for high- and low-grade gliomas and to address such issues as pseudoresponse and nonenhancing tumor progression from antiangiogenic therapies, and pseudoprogression from radiochemotherapy. RANO has expanded to include working groups that are focused on other tumors, including brain metastases, leptomeningeal metastases, spine tumors, pediatric brain tumors, and meningiomas, as well as other clinical trial end points, such as clinical outcomes assessments, seizures, corticosteroid use, and positron emission tomography imaging. In an effort to standardize the measurement of neurologic function for clinical assessment, the Neurologic Assessment in Neuro-Oncology scale was drafted. Born out of a workshop conducted by the Jumpstarting Brain Tumor Drug Development Coalition and the US Food and Drug Administration, a standardized brain tumor imaging protocol now exists to reduce variability and improve reliability. Efforts by RANO have been widely accepted and are increasingly being used in neuro-oncology trials, although additional refinements will be needed.
Collapse
Affiliation(s)
- Patrick Y. Wen
- Patrick Y. Wen and Eudocia Q. Lee, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Susan M. Chang, University of California, San Francisco, San Francisco, CA; Michael A. Vogelbaum, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Martin J. Van den Bent, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands; and David R. Macdonald, London Regional Cancer Program, Western University, London, Ontario, Canada
| | - Susan M. Chang
- Patrick Y. Wen and Eudocia Q. Lee, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Susan M. Chang, University of California, San Francisco, San Francisco, CA; Michael A. Vogelbaum, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Martin J. Van den Bent, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands; and David R. Macdonald, London Regional Cancer Program, Western University, London, Ontario, Canada
| | - Martin J. Van den Bent
- Patrick Y. Wen and Eudocia Q. Lee, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Susan M. Chang, University of California, San Francisco, San Francisco, CA; Michael A. Vogelbaum, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Martin J. Van den Bent, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands; and David R. Macdonald, London Regional Cancer Program, Western University, London, Ontario, Canada
| | - Michael A. Vogelbaum
- Patrick Y. Wen and Eudocia Q. Lee, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Susan M. Chang, University of California, San Francisco, San Francisco, CA; Michael A. Vogelbaum, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Martin J. Van den Bent, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands; and David R. Macdonald, London Regional Cancer Program, Western University, London, Ontario, Canada
| | - David R. Macdonald
- Patrick Y. Wen and Eudocia Q. Lee, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Susan M. Chang, University of California, San Francisco, San Francisco, CA; Michael A. Vogelbaum, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Martin J. Van den Bent, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands; and David R. Macdonald, London Regional Cancer Program, Western University, London, Ontario, Canada
| | - Eudocia Q. Lee
- Patrick Y. Wen and Eudocia Q. Lee, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Susan M. Chang, University of California, San Francisco, San Francisco, CA; Michael A. Vogelbaum, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Martin J. Van den Bent, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands; and David R. Macdonald, London Regional Cancer Program, Western University, London, Ontario, Canada
| |
Collapse
|
42
|
Packer RJ, Pfister S, Bouffet E, Avery R, Bandopadhayay P, Bornhorst M, Bowers DC, Ellison D, Fangusaro J, Foreman N, Fouladi M, Gajjar A, Haas-Kogan D, Hawkins C, Ho CY, Hwang E, Jabado N, Kilburn LB, Lassaletta A, Ligon KL, Massimino M, Meeteren SV, Mueller S, Nicolaides T, Perilongo G, Tabori U, Vezina G, Warren K, Witt O, Zhu Y, Jones DT, Kieran M. Pediatric low-grade gliomas: implications of the biologic era. Neuro Oncol 2017; 19:750-761. [PMID: 27683733 PMCID: PMC5464436 DOI: 10.1093/neuonc/now209] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
For the past decade, it has been recognized that pediatric low-grade gliomas (LGGs) and glial-neuronal tumors carry distinct molecular alterations with resultant aberrant intracellular signaling in the Ras-mitogen-activated protein kinase pathway. The conclusions and recommendations of a consensus conference of how best to integrate the growing body of molecular genetic information into tumor classifications and, more importantly, for future treatment of pediatric LGGs are summarized here. There is uniform agreement that molecular characterization must be incorporated into classification and is increasingly critical for appropriate management. Molecular-targeted therapies should be integrated expeditiously, but also carefully into the management of these tumors and success measured not only by radiographic responses or stability, but also by functional outcomes. These trials need to be carried out with the caveat that the long-term impact of molecularly targeted therapy on the developing nervous system, especially with long duration treatment, is essentially unknown.
Collapse
Affiliation(s)
- Roger J Packer
- Center for Neuroscience and Behavioral Medicine, Washington, District of Columbia, USA
- Gilbert Family Neurofibromatosis Institute, Washington, District of Columbia, USA
- Brain Tumor Institute, Washington, District of Columbia, USA
| | - Stephan Pfister
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Eric Bouffet
- Paediatric Neuro-Oncology Program, Research Institute and The Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Robert Avery
- Center for Neuroscience and Behavioral Medicine, Washington, District of Columbia, USA
- Gilbert Family Neurofibromatosis Institute, Washington, District of Columbia, USA
- Brain Tumor Institute, Washington, District of Columbia, USA
| | - Pratiti Bandopadhayay
- Department of Pediatrics, Brigham and Women's Hospital, Harvard Medical School, and the Broad Institute, Dana-Farber/Boston Children's Cancer and Blood Disorders Centre, Boston, Massachusetts, USA
| | - Miriam Bornhorst
- Center for Neuroscience and Behavioral Medicine, Washington, District of Columbia, USA
- Gilbert Family Neurofibromatosis Institute, Washington, District of Columbia, USA
- Brain Tumor Institute, Washington, District of Columbia, USA
- Center for Cancer and Immunology Research, Washington, District of Columbia, USA
| | - Daniel C Bowers
- Department of Pediatrics, UT Southwestern Medical School, Dallas, Texas, USA
| | - David Ellison
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee. USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jason Fangusaro
- Ann and Robert H. Lurie Children's Hospital of Chicago Department of Pediatric Hematology/Oncology and Stem Cell Transplantation, University of Colorado, Aurora, Colorado, USA
| | - Nicholas Foreman
- Northwestern Feinberg School of Medicine, Chicago, Illinois; Children's Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| | - Maryam Fouladi
- Brain Tumor Center, Brain Tumor Translational Research, UC Department of Pediatrics, Cincinnati, Ohio, USA
| | - Amar Gajjar
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Daphne Haas-Kogan
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, and the Broad Institute, Dana-Farber/Boston Children's Cancer and Blood Disorders Centre, Boston, Massachusetts, USA
| | - Cynthia Hawkins
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee. USA
| | - Cheng-Ying Ho
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Eugene Hwang
- Center for Neuroscience and Behavioral Medicine, Washington, District of Columbia, USA
- Brain Tumor Institute, Washington, District of Columbia, USA
- Center for Cancer and Immunology Research, Washington, District of Columbia, USA
| | - Nada Jabado
- Ann and Robert H. Lurie Children's Hospital of Chicago Department of Pediatric Hematology/Oncology and Stem Cell Transplantation, University of Colorado, Aurora, Colorado, USA
| | - Lindsay B Kilburn
- Brain Tumor Institute, Washington, District of Columbia, USA
- Center for Cancer and Immunology Research, Washington, District of Columbia, USA
| | - Alvaro Lassaletta
- Northwestern Feinberg School of Medicine, Chicago, Illinois; Children's Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| | - Keith L Ligon
- Brain Tumor Center, Brain Tumor Translational Research, UC Department of Pediatrics, Cincinnati, Ohio, USA
- Brain Tumor Center, Brain Tumor Translational Research, UC Department of Pediatrics, Cincinnati, Ohio, USA
| | - Maura Massimino
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, and the Broad Institute, Dana-Farber/Boston Children's Cancer and Blood Disorders Centre, Boston, Massachusetts, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, and the Broad Institute, Dana-Farber/Boston Children's Cancer and Blood Disorders Centre, Boston, Massachusetts, USA
| | | | - Sabine Mueller
- Department of Neurology, Pediatrics and Neurosurgery, University of California San Francisco, San Francisco, California, USA
| | - Theo Nicolaides
- Department of Neurology, Pediatrics and Neurosurgery, University of California San Francisco, San Francisco, California, USA
| | - Giorgio Perilongo
- Department of Woman's and Child's Health, University of Padua, Padua, Italy
| | - Uri Tabori
- Division of Haematology/Oncology, Research Institute and The Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Gilbert Vezina
- Gilbert Family Neurofibromatosis Institute, Washington, District of Columbia, USA
- Brain Tumor Institute, Washington, District of Columbia, USA
- Division of Neuroradiology, Washington, District of Columbia, USA
| | - Katherine Warren
- National Cancer Institute, Pediatric Oncology and Neuro-Oncology Branches, Bethesda, Maryland, USA
| | - Olaf Witt
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yuan Zhu
- Center for Neuroscience and Behavioral Medicine, Washington, District of Columbia, USA
- Gilbert Family Neurofibromatosis Institute, Washington, District of Columbia, USA
- Center for Cancer and Immunology Research, Washington, District of Columbia, USA
| | - David T Jones
- Division of Pediatric Neuro-oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mark Kieran
- Brain Tumor Center, Brain Tumor Translational Research, UC Department of Pediatrics, Cincinnati, Ohio, USA
| |
Collapse
|
43
|
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
|
44
|
Svolos P, Reddick WE, Edwards A, Sykes A, Li Y, Glass JO, Patay Z. Measurable Supratentorial White Matter Volume Changes in Patients with Diffuse Intrinsic Pontine Glioma Treated with an Anti-Vascular Endothelial Growth Factor Agent, Steroids, and Radiation. AJNR Am J Neuroradiol 2017; 38:1235-1241. [PMID: 28428205 DOI: 10.3174/ajnr.a5159] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 01/26/2017] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Assessing the response to treatment in infiltrative brain tumors by using lesion volume-based response criteria is challenging. We hypothesized that in such tumors, volume measurements alone may not accurately capture changes in actual tumor burden during treatment. We longitudinally evaluated volume changes in both normal-appearing supratentorial white matter and the brain stem lesions in patients treated for diffuse intrinsic pontine glioma to determine to what extent adjuvant systemic therapies may skew the accuracy of tumor response assessments based on volumetric analysis. MATERIALS AND METHODS The anatomic MR imaging and diffusion tensor imaging data of 26 patients with diffuse intrinsic pontine glioma were retrospectively analyzed. Treatment included conformal radiation therapy in conjunction with vandetanib and dexamethasone. Volumetric and diffusion data were analyzed with time, and differences between time points were evaluated statistically. RESULTS Normalized brain stem lesion volume decreased during combined treatment (slope = -0.222, P < .001) and increased shortly after completion of radiation therapy (slope = 0.422, P < .001). Supratentorial white matter volume steadily and significantly decreased with time (slope = -0.057, P < .001). CONCLUSIONS Longitudinal changes in brain stem lesion volume are robust; less pronounced but measurable changes occur in the supratentorial white matter. Volume changes in nonirradiated supratentorial white matter during the disease course reflect the effects of systemic medication on the water homeostasis of normal parenchyma. Our data suggest that adjuvant nontumor-targeted therapies may have a more substantial effect on lesion volume changes than previously thought; hence, an apparent volume decrease in infiltrative tumors receiving combined therapies may lead to overestimation of the actual response and tumor control.
Collapse
Affiliation(s)
- P Svolos
- From the Departments of Diagnostic Imaging (P.S., W.E.R., A.E., J.O.G., Z.P.)
| | - W E Reddick
- From the Departments of Diagnostic Imaging (P.S., W.E.R., A.E., J.O.G., Z.P.)
| | - A Edwards
- From the Departments of Diagnostic Imaging (P.S., W.E.R., A.E., J.O.G., Z.P.)
| | - A Sykes
- Biostatistics (A.S., Y.L.), St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Y Li
- Biostatistics (A.S., Y.L.), St. Jude Children's Research Hospital, Memphis, Tennessee
| | - J O Glass
- From the Departments of Diagnostic Imaging (P.S., W.E.R., A.E., J.O.G., Z.P.)
| | - Z Patay
- From the Departments of Diagnostic Imaging (P.S., W.E.R., A.E., J.O.G., Z.P.)
| |
Collapse
|
45
|
Carceller F, Jerome NP, Miyazaki K, Collins DJ, Orton MR, d'Arcy JA, Wallace T, Moreno L, Pearson ADJ, Zacharoulis S, Leach MO, Marshall LV, Koh DM. Feasibility and applicability of diffusion-weighted and dynamic contrast-enhanced magnetic resonance imaging in routine assessments of children with high-grade gliomas. Pediatr Blood Cancer 2017; 64:279-283. [PMID: 27615273 DOI: 10.1002/pbc.26216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 07/26/2016] [Accepted: 07/26/2016] [Indexed: 12/28/2022]
Abstract
Diffusion-weighted (DW) and dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI) have been used as imaging biomarkers in adults with high-grade gliomas (HGGs). We incorporated free-breathing DW-MRI and DCE-MRI, at a single time point, in the routine follow-up of five children (median age 9 years, range 8-15) with histologically confirmed HGG within a prospective imaging study. It was feasible to incorporate DW-MRI and DCE-MRI in routine assessments of children with HGG. DW and DCE parameters were repeatable in paediatric HGG. Higher median ADC100-1000 significantly correlated with longer survival in our sample.
Collapse
Affiliation(s)
- Fernando Carceller
- Paediatric & Adolescent Drug Development Team, Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Neil P Jerome
- Cancer Research UK Cancer Imaging Centre, The Institute of Cancer Research, London, United Kingdom
| | - Keiko Miyazaki
- Cancer Research UK Cancer Imaging Centre, The Institute of Cancer Research, London, United Kingdom
| | - David J Collins
- Cancer Research UK Cancer Imaging Centre, The Institute of Cancer Research, London, United Kingdom
| | - Matthew R Orton
- Cancer Research UK Cancer Imaging Centre, The Institute of Cancer Research, London, United Kingdom
| | - James A d'Arcy
- Cancer Research UK Cancer Imaging Centre, The Institute of Cancer Research, London, United Kingdom
| | - Toni Wallace
- Radiology Department, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Lucas Moreno
- Paediatric & Adolescent Drug Development Team, Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
- Clinical Trials Unit, Paediatric Oncology Department, Hospital Niño Jesús, Madrid, Spain
| | - Andrew D J Pearson
- Paediatric & Adolescent Drug Development Team, Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Stergios Zacharoulis
- Paediatric & Adolescent Drug Development Team, Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Martin O Leach
- Cancer Research UK Cancer Imaging Centre, The Institute of Cancer Research, London, United Kingdom
| | - Lynley V Marshall
- Paediatric & Adolescent Drug Development Team, Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Dow-Mu Koh
- Radiology Department, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
46
|
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
|
47
|
Huang RY, Wen PY. Response Assessment in Neuro-Oncology Criteria and Clinical Endpoints. Magn Reson Imaging Clin N Am 2016; 24:705-718. [PMID: 27742111 DOI: 10.1016/j.mric.2016.06.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The Response Assessment in Neuro-Oncology (RANO) Working Group is an international multidisciplinary group whose goal is to improve response criteria and define endpoints for neuro-oncology trials. The RANO criteria for high-grade gliomas attempt to address the issues of pseudoprogression, pseudoresponse, and nonenhancing tumor progression. Incorporation of advanced MR imaging may eventually help improve the ability of these criteria to define enhancing and nonenhancing disease better. The RANO group has also developed criteria for neurologic response and evaluation of patients receiving immunologic therapies. RANO criteria have been developed for brain metastases and are in progress for meningiomas, leptomeningeal disease, spinal tumors, and pediatric tumors.
Collapse
Affiliation(s)
- Raymond Y Huang
- Division of Neuroradiology, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| | - Patrick Y Wen
- Division of Neuro-Oncology, Department of Neurology, Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
48
|
Carceller F, Bautista FJ, Fowkes LA, Marshall LV, Sirvent SI, Chisholm JC, Pearson ADJ, Koh DM, Moreno L. Response Assessment in Paediatric Phase I Trials According to RECIST Guidelines: Survival Outcomes, Patterns of Progression and Relevance of Changes in Tumour Measurements. Pediatr Blood Cancer 2016; 63:1400-6. [PMID: 27149049 DOI: 10.1002/pbc.26039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 04/04/2016] [Indexed: 11/06/2022]
Abstract
INTRODUCTION RECIST guidelines constitute the reference for radiological response assessment in most paediatric trials of anticancer agents. However, these criteria have not been validated in children. We evaluated the outcomes and patterns of progression of children/adolescents enrolled in phase I trials in two paediatric drug development units. METHODS Patients aged ≤21 assessed with RECIST (v1.0 or v1.1) were eligible. Clinico-radiological data were analysed using Mann-Whitney U and log-rank tests to correlate response categories and sum of longest diameters (SLD) with time-to-event variables and overall survival (OS). RESULTS Sixty-one patients (71 enrolments) were evaluated; median age: 12.7 years (range, 3.1-20.9). Overall, 7% achieved complete/partial response (n = 5) and 31% disease stabilisation (n = 22). Median (95% CI) OS (in months) was 29.1 (27.6-30.6) with complete/partial response, 8.9 (2.0-15.8) with stable disease and 2.8 (2.3-3.3) with disease progression (P < 0.001); 32.6% patients with measurable disease presented exclusive progression of existing non-target lesions and/or new lesions. The change in SLD at best response showed a linear correlation with duration of response (r = -0.605; P = 0.004) and time on trial (r = -0.61; P = 0.003), but the change in SLD at progression did not correlate with time to progression (r = -0.219; P = 0.206). CONCLUSIONS Response assessment according to RECIST correlated with OS in children/adolescents treated on phase I trials. The reduction in SLD at best response correlated with more prolonged responses. Tumour size did not constitute an optimal method to assess disease progression in one third of patients with measurable disease. Further refinement of current response assessment guidelines will enable the development of paediatric-specific radiological criteria.
Collapse
Affiliation(s)
- Fernando Carceller
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Francisco J Bautista
- Paediatric Oncology Department, Clinical Trials Unit, Hospital Niño Jesús, Madrid, Spain
| | - Lucy A Fowkes
- Radiology Department, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Lynley V Marshall
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Sara I Sirvent
- Paediatric Radiology Department, Hospital Niño Jesús, Madrid, Spain
| | - Julia C Chisholm
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Andrew D J Pearson
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Dow-Mu Koh
- Radiology Department, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Lucas Moreno
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
- Paediatric Oncology Department, Clinical Trials Unit, Hospital Niño Jesús, Madrid, Spain
- Clinical Research Programme, CNIO (Spanish National Cancer Research Centre), Madrid, Spain
| |
Collapse
|
49
|
Carceller F, Fowkes LA, Khabra K, Moreno L, Saran F, Burford A, Mackay A, Jones DTW, Hovestadt V, Marshall LV, Vaidya S, Mandeville H, Jerome N, Bridges LR, Laxton R, Al-Sarraj S, Pfister SM, Leach MO, Pearson ADJ, Jones C, Koh DM, Zacharoulis S. Pseudoprogression in children, adolescents and young adults with non-brainstem high grade glioma and diffuse intrinsic pontine glioma. J Neurooncol 2016; 129:109-21. [PMID: 27180091 DOI: 10.1007/s11060-016-2151-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 05/07/2016] [Indexed: 02/01/2023]
Abstract
Pseudoprogression (PsP) is a treatment-related phenomenon which hinders response interpretation. Its prevalence and clinical impact have not been evaluated in children/adolescents. We assessed the characteristics, risk factors and prognosis of PsP in children/adolescents and young-adults diagnosed with non-brainstem high grade gliomas (HGG) and diffuse intrinsic pontine gliomas (DIPG). Patients aged 1-21 years diagnosed with HGG or DIPG between 1995 and 2012 who had completed radiotherapy were eligible. PsP was assessed according to study-specific criteria and correlated with first-line treatment, molecular biomarkers and survival. Ninety-one patients (47 HGG, 44 DIPG) were evaluable. Median age: 10 years (range, 2-20). Eleven episodes of PsP were observed in 10 patients (4 HGG, 6 DIPG). Rates of PsP: 8.5 % (HGG); 13.6 % (DIPG). Two episodes of PsP were based on clinical findings alone; nine episodes had concurrent radiological changes: increased size of lesions (n = 5), new focal enhancement (n = 4). Temozolomide, MGMT methylation or H3F3A mutations were not found to be associated with increased occurrence of PsP. For HGG, 1-year progression-free survival (PFS) was 41.9 % no-PsP versus 100 % PsP (p = 0.041); differences in 1-year overall survival (OS) were not significant. For DIPG, differences in 1-year PFS and OS were not statistically significant. Hazard ratio (95 %CI) of PsP for OS was 0.551 (0.168-1.803; p = 0.325) in HGG; and 0.308 (0.107-0.882; p = 0.028) in DIPG. PsP occurred in both pediatric HGG and DIPG patients at a comparable rate to adult HGG. PsP was associated with improved 1-yr PFS in HGG patients. PsP had a protective effect upon OS in DIPG patients.
Collapse
Affiliation(s)
- Fernando Carceller
- Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
- Division of Clinical Studies and Cancer Therapeutics, Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK.
| | - Lucy A Fowkes
- Department of Radiology, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, SM2 5PT, UK
| | - Komel Khabra
- Research Data Management and Statistics Unit, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, SM2 5PT, UK
| | - Lucas Moreno
- Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK
- Division of Clinical Studies and Cancer Therapeutics, Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
- Clinical Research Unit - Pediatric Phase I-II Clinical Trials, Pediatric Oncology-Hematology Service, Hospital Niño Jesús, Av. de Menéndez Pelayo, num 65, 28009, Madrid, Spain
| | - Frank Saran
- Department of Neuro Oncology, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, SM2 5PT, UK
| | - Anna Burford
- Division of Clinical Studies and Cancer Therapeutics, Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
- Division of Molecular Pathology, Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
| | - Alan Mackay
- Division of Clinical Studies and Cancer Therapeutics, Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
- Division of Molecular Pathology, Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
| | - David T W Jones
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69121, Heidelberg, Germany
| | - Volker Hovestadt
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69121, Heidelberg, Germany
| | - Lynley V Marshall
- Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK
- Division of Clinical Studies and Cancer Therapeutics, Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
- Division of Molecular Pathology, Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
| | - Sucheta Vaidya
- Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK
- Division of Clinical Studies and Cancer Therapeutics, Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
| | - Henry Mandeville
- Department of Radiotherapy, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, SM2 5PT, UK
| | - Neil Jerome
- CRUK Cancer Imaging Centre, Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
| | - Leslie R Bridges
- Department of Cellular Pathology, St George's Hospital, Blackshaw Road, London, SW17 0QT, UK
| | - Ross Laxton
- Department of Clinical Neuropathology, King's College Hospital, Denmark Hill, London, SE5 9RS, UK
| | - Safa Al-Sarraj
- Department of Clinical Neuropathology, King's College Hospital, Denmark Hill, London, SE5 9RS, UK
| | - Stefan M Pfister
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69121, Heidelberg, Germany
- Department of Pediatric Oncology and Hematology, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 672, 69120, Heidelberg, Germany
| | - Martin O Leach
- CRUK Cancer Imaging Centre, Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
| | - Andrew D J Pearson
- Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK
- Division of Clinical Studies and Cancer Therapeutics, Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
| | - Chris Jones
- Division of Clinical Studies and Cancer Therapeutics, Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
- Division of Molecular Pathology, Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
| | - Dow-Mu Koh
- Department of Radiology, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, SM2 5PT, UK
| | - Stergios Zacharoulis
- Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK
- Division of Clinical Studies and Cancer Therapeutics, Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
| |
Collapse
|
50
|
Discrepant longitudinal volumetric and metabolic evolution of diffuse intrinsic Pontine gliomas during treatment: implications for current response assessment strategies. Neuroradiology 2016; 58:1027-1034. [PMID: 27438806 DOI: 10.1007/s00234-016-1724-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/04/2016] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Based on clinical observations, we hypothesized that in infiltrative high-grade brainstem neoplasms, such as diffuse intrinsic pontine glioma (DIPG), longitudinal metabolic evaluation of the tumor by magnetic resonance spectroscopy (MRS) may be more accurate than volumetric data for monitoring the tumor's biological evolution during standard treatment. METHODS We evaluated longitudinal MRS data and corresponding tumor volumes of 31 children with DIPG. We statistically analyzed correlations between tumor volume and ratios of Cho/NAA, Cho/Cr, and NAA/Cr at key time points during the course of the disease through the end of the progression-free survival period. RESULTS By the end of RT, tumor volume had significantly decreased from the baseline (P < .0001) and remained decreased through the last available follow-up magnetic resonance imaging study (P = .007632). However, the metabolic profile of the tumor tissue (Cho/Cr, NAA/Cr, and Cho/NAA ratios) did not change significantly over time. CONCLUSION Our data show that longitudinal tumor volume and metabolic profile changes are dissociated in patients with DIPG during progression-free survival. Volume changes, therefore, may not accurately reflect treatment-related changes in tumor burden. This study adds to the existing body of evidence that the value of conventional MRI metrics, including volumetric data, needs to be reevaluated critically and, in infiltrative tumors in particular, may not be useful as study end-points in clinical trials. We submit that advanced quantitative MRI data, including robust, MRS-based metabolic ratios and diffusion and perfusion metrics, may be better surrogate markers of key end-points in clinical trials.
Collapse
|