1
|
Ahmed S, Abdullah M, Khan MAA, Resham S, Qureshi BM, Mushtaq N. The trends in diagnosis, management, and care of patients with diffuse intrinsic pontine gliomas: Perspectives from a tertiary care hospital of pakistan. Childs Nerv Syst 2024; 40:3537-3544. [PMID: 39349775 DOI: 10.1007/s00381-024-06637-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 09/24/2024] [Indexed: 11/06/2024]
Abstract
BACKGROUND Diffuse intrinsic pontine glioma (DIPG) primarily affects pediatric patients. Data on the global incidence of DIPG remain sparse, especially in South Asia and low-middle-income countries like Pakistan. METHODS After exemption from the Ethics Review Committee, a retrospective study was conducted. Records of patients with DIPG at the Aga Khan Hospital in Karachi, from January 2010 to December 2022, were reviewed. RESULTS A total of 35 pediatric patients were managed for DIPG. The median age of the patients was 9, with 19 (54.3%) males and 16 (45.7%) females. Cranial nerve palsies were the most common complaint and were present in 19 (54.3%) patients, followed by headaches in 18 (51.4%), long tract signs in 14 (40%), ataxia/cerebellar symptoms in 14 (40%), and seizures in 5 (14.3%). MRI was the primary diagnostic tool, used alone or with CT in 32 (94.1%) patients; CT alone was used in only 2 (5.7%) patients. Biopsy was performed in 10 (28.6%) patients. Primary radiation therapy was administered to 14 (40%) patients with 5400 cGy in 30 fractions. All these patients received steroids while none of them received reirradiation. VP shunt surgery for hydrocephalus was performed in 9 (25.7%) patients. Over half (54.3%) refused treatment post-diagnosis, and 71.4% were lost to follow-up. CONCLUSION Providing timely, quality multi-disciplinary care to DIPG patients within resource constraints remains challenging in Pakistan. However, recent developments show promise for improving DIPG care in the country.
Collapse
Affiliation(s)
- Salaar Ahmed
- Medical College, Aga Khan University, Karachi, 74800, Pakistan
| | | | | | - Shahzadi Resham
- Department of Oncology, Aga Khan University Hospital, Karachi, 74800, Pakistan
| | | | - Naureen Mushtaq
- Department of Oncology, Aga Khan University Hospital, Karachi, 74800, Pakistan.
| |
Collapse
|
2
|
Huangfu L, Zha B, Li P, Wang L, Liu X, Cui H, Li Y, Wu J, Shi S, Yang Y, Sun X, Gao S, Li H, Yang D, Zheng Y. A phase I clinical trial of sonodynamic therapy combined with radiotherapy for brainstem gliomas. Int J Cancer 2024. [PMID: 39377640 DOI: 10.1002/ijc.35218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/09/2024]
Abstract
Brainstem gliomas (BSGs) are a class of clinically refractory malignant tumors for which there is no uniform and effective treatment protocol. Ultrasound and radiation can activate hematoporphyrin and produce sonodynamic and radiodynamic effects to kill cancer cells. Therefore, we conducted the first phase I clinical trial of sonodynamic therapy (SDT) combined with radiotherapy (RT) for the treatment of BSGs to verify its safety and efficacy. We conducted a study of SDT combined with RT in 11 patients with BSGs who received SDT and RT after hematoporphyrin administration. Magnetic resonance imaging was performed during this period to assess the tumor, and adverse events were recorded. All adverse events recorded were grade 1-2; no grade 3 or more serious adverse events were observed. Treatment was well tolerated, and no dose-limiting toxicities were observed. There were no treatment-related deaths during the course of treatment. 8 of 11 patients (72.7%) maintained stable disease, 2 (18.2%) achieved partial response, and the tumors were still shrinking as of the last follow-up date. The median progression-free survival (PFS) for patients was 9.2 (95% confidence interval [CI] 6.2-12.2) months, and the median overall survival (OS) was 11.7 (95% CI 9.6-13.8) months. Therefore, SDT combined with RT has a favorable safety and feasibility and shows a preliminary high therapeutic potential.
Collapse
Affiliation(s)
- Linkuan Huangfu
- Department of Radiotherapy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Boya Zha
- Department of Radiotherapy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Peihong Li
- Department of Radiotherapy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Long Wang
- Department of Radiotherapy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Xiaohao Liu
- Department of Radiotherapy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Haiyang Cui
- Department of Radiotherapy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Yuxin Li
- Department of Radiotherapy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Jingjing Wu
- Department of Radiotherapy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Shuling Shi
- Department of Radiotherapy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Yuchuan Yang
- Department of Radiotherapy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Xiaocong Sun
- Department of Radiotherapy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Shibo Gao
- Department of Radiotherapy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Huizhen Li
- Department of Radiotherapy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Daoke Yang
- Department of Radiotherapy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Yingjuan Zheng
- Department of Radiotherapy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
- Department of Hyperthermia and Photodynamic Therapy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| |
Collapse
|
3
|
Kubendiran L. Letter to the Editor: "The relationship between imaging features, therapeutic response, and overall survival in pediatric diffuse intrinsic pontine glioma". Neurosurg Rev 2024; 47:427. [PMID: 39141137 DOI: 10.1007/s10143-024-02676-7] [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: 07/29/2024] [Revised: 07/29/2024] [Accepted: 08/10/2024] [Indexed: 08/15/2024]
Abstract
This letter to the editor discusses the findings of Yu et al. (2024), which highlight the prognostic significance of volumetric assessments over cross-product measurements in pediatric diffuse intrinsic pontine glioma (DIPG). The study's methodology enhances precision in monitoring therapeutic responses, offering insights into treatment adjustments based on detailed imaging features. Emphasizing the value of volumetric MRI, this letter suggests its potential to improve surgical planning and therapeutic strategies, thereby optimizing patient management. This approach could revolutionize treatment paradigms, emphasizing personalized care through advanced imaging techniques.
Collapse
Affiliation(s)
- Lavanya Kubendiran
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu, India.
| |
Collapse
|
4
|
Kazerooni AF, Khalili N, Liu X, Haldar D, Jiang Z, Anwar SM, Albrecht J, Adewole M, Anazodo U, Anderson H, Bagheri S, Baid U, Bergquist T, Borja AJ, Calabrese E, Chung V, Conte GM, Dako F, Eddy J, Ezhov I, Familiar A, Farahani K, Haldar S, Iglesias JE, Janas A, Johansen E, Jones BV, Kofler F, LaBella D, Lai HA, Van Leemput K, Li HB, Maleki N, McAllister AS, Meier Z, Menze B, Moawad AW, Nandolia KK, Pavaine J, Piraud M, Poussaint T, Prabhu SP, Reitman Z, Rodriguez A, Rudie JD, Shaikh IS, Shah LM, Sheth N, Shinohara RT, Tu W, Viswanathan K, Wang C, Ware JB, Wiestler B, Wiggins W, Zapaishchykova A, Aboian M, Bornhorst M, de Blank P, Deutsch M, Fouladi M, Hoffman L, Kann B, Lazow M, Mikael L, Nabavizadeh A, Packer R, Resnick A, Rood B, Vossough A, Bakas S, Linguraru MG. The Brain Tumor Segmentation (BraTS) Challenge 2023: Focus on Pediatrics (CBTN-CONNECT-DIPGR-ASNR-MICCAI BraTS-PEDs). ARXIV 2024:arXiv:2305.17033v7. [PMID: 37292481 PMCID: PMC10246083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Pediatric tumors of the central nervous system are the most common cause of cancer-related death in children. The five-year survival rate for high-grade gliomas in children is less than 20%. Due to their rarity, the diagnosis of these entities is often delayed, their treatment is mainly based on historic treatment concepts, and clinical trials require multi-institutional collaborations. The MICCAI Brain Tumor Segmentation (BraTS) Challenge is a landmark community benchmark event with a successful history of 12 years of resource creation for the segmentation and analysis of adult glioma. Here we present the CBTN-CONNECT-DIPGR-ASNR-MICCAI BraTS-PEDs 2023 challenge, which represents the first BraTS challenge focused on pediatric brain tumors with data acquired across multiple international consortia dedicated to pediatric neuro-oncology and clinical trials. The BraTS-PEDs 2023 challenge focuses on benchmarking the development of volumentric segmentation algorithms for pediatric brain glioma through standardized quantitative performance evaluation metrics utilized across the BraTS 2023 cluster of challenges. Models gaining knowledge from the BraTS-PEDs multi-parametric structural MRI (mpMRI) training data will be evaluated on separate validation and unseen test mpMRI dataof high-grade pediatric glioma. The CBTN-CONNECT-DIPGR-ASNR-MICCAI BraTS-PEDs 2023 challenge brings together clinicians and AI/imaging scientists to lead to faster development of automated segmentation techniques that could benefit clinical trials, and ultimately the care of children with brain tumors.
Collapse
Affiliation(s)
- Anahita Fathi Kazerooni
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA
- Center for AI & Data Science for Integrated Diagnostics (AI2D) and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
| | - Nastaran Khalili
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Xinyang Liu
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington DC, USA
| | - Debanjan Haldar
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Thomas Jefferson University Hospital, PA, USA
| | - Zhifan Jiang
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington DC, USA
| | - Syed Muhammed Anwar
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington DC, USA
| | | | - Maruf Adewole
- Medical Artificial Intelligence (MAI) Lab, Crestview Radiology, Lagos, Nigeria
| | - Udunna Anazodo
- Montreal Neurological Institute (MNI), McGill University, Montreal, QC, Canada
| | - Hannah Anderson
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sina Bagheri
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ujjwal Baid
- Center for AI & Data Science for Integrated Diagnostics (AI2D) and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Austin J. Borja
- Department of Neurosurgery at the University of Southern California, CA, USA
| | - Evan Calabrese
- Department of Radiology, Duke University Medical Center, USA
| | | | | | - Farouk Dako
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Ivan Ezhov
- Department of Informatics, Technical University Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Germany
| | - Ariana Familiar
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Keyvan Farahani
- Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shuvanjan Haldar
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Biomedical Engineering Rutgers University, New Brunswick, NJ, USA
| | - Juan Eugenio Iglesias
- Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | | | - Elaine Johansen
- PrecisionFDA, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | | | | | - Dominic LaBella
- Department of Radiation Oncology, Duke University Medical Center, USA
| | - Hollie Anne Lai
- Department of Radiology, Children’s Health Orange County, CA, USA
| | - Koen Van Leemput
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Denmark
| | - Hongwei Bran Li
- Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | | | | | | | - Bjoern Menze
- Biomedical Image Analysis & Machine Learning, Department of Quantitative Biomedicine, University of Zurich, Switzerland
- Department of Neuroradiology, Technical University of Munich, Munich, Germany
| | | | - Khanak K Nandolia
- Department of Diagnostic and Interventional Radiology, All India Institute of Medical Sciences, Rishikesh, India
| | - Julija Pavaine
- Department of Paediatric Radiology, Royal Manchester Children’s Hospital, Manchester University Hospitals NHS Foundation Trust, University of Manchester, Manchester, UK
- Division of Informatics, Imaging & Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | | | - Tina Poussaint
- Dana-Farber Brigham Cancer Center & Boston Children’s Hospital, Boston, MA, USA
| | - Sanjay P Prabhu
- Division of Neuroradiology, Department of Radiology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Zachary Reitman
- Department of Radiation Oncology, Duke University Medical Center, USA
| | - Andres Rodriguez
- Department of Diagnostic & Interventional Imaging, Neuroradiology section. The University of Texas Health Sciences Center at Houston
| | | | | | | | - Nakul Sheth
- Department of Radiology, The Children’s Hospital of Philadelphia, PA, USA
| | - Russel Taki Shinohara
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, USA
| | - Wenxin Tu
- College of Arts and Sciences, University of Pennsylvania, PA, USA
| | - Karthik Viswanathan
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Chunhao Wang
- Department of Radiation Oncology, Duke University Medical Center, USA
| | - Jeffrey B Ware
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benedikt Wiestler
- Department of Neuroradiology, Technical University of Munich, Munich, Germany
| | - Walter Wiggins
- Department of Radiology, Duke University Medical Center, USA
| | - Anna Zapaishchykova
- Dana-Farber Brigham Cancer Center & Boston Children’s Hospital, Boston, MA, USA
| | | | - Miriam Bornhorst
- Brain Tumor Institute, Children’s National Hospital, Washington, DC, USA
| | - Peter de Blank
- Brain Tumor Center, Cincinnati Children’s Hospital, Cincinnati, OH, USA
| | - Michelle Deutsch
- Pediatric Neuro-Oncology Program, Nationwide Children’s Hospital, Columbus, OH, US
| | - Maryam Fouladi
- Pediatric Neuro-Oncology Program, Nationwide Children’s Hospital, Columbus, OH, US
| | - Lindsey Hoffman
- Center for Cancer and Blood Disorders, Phoenix Children’s Hospital, Phoenix, AZ, US
| | - Benjamin Kann
- Dana-Farber Brigham Cancer Center & Boston Children’s Hospital, Boston, MA, USA
| | - Margot Lazow
- Pediatric Neuro-Oncology Program, Nationwide Children’s Hospital, Columbus, OH, US
| | - Leonie Mikael
- Pediatric Neuro-Oncology Program, Nationwide Children’s Hospital, Columbus, OH, US
| | - Ali Nabavizadeh
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Roger Packer
- Brain Tumor Institute, Children’s National Hospital, Washington, DC, USA
| | - Adam Resnick
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Brian Rood
- Center for Cancer and Blood Disorders, Children’s National Hospital, Washington, DC, USA
| | - Arastoo Vossough
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Radiology, The Children’s Hospital of Philadelphia, PA, USA
| | - Spyridon Bakas
- Center for AI & Data Science for Integrated Diagnostics (AI2D) and Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marius George Linguraru
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington DC, USA
- Departments of Radiology and Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
5
|
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
|
6
|
Mueller S, Kline C, Franson A, van der Lugt J, Prados M, Waszak SM, Plasschaert SLA, Molinaro AM, Koschmann C, Nazarian J. Rational combination platform trial design for children and young adults with diffuse midline glioma: A report from PNOC. Neuro Oncol 2024; 26:S125-S135. [PMID: 38124481 PMCID: PMC11066905 DOI: 10.1093/neuonc/noad181] [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: 05/23/2023] [Indexed: 12/23/2023] Open
Abstract
Background Diffuse midline glioma (DMG) is a devastating pediatric brain tumor unresponsive to hundreds of clinical trials. Approximately 80% of DMGs harbor H3K27M oncohistones, which reprogram the epigenome to increase the metabolic profile of the tumor cells. Methods We have previously shown preclinical efficacy of targeting both oxidative phosphorylation and glycolysis through treatment with ONC201, which activates the mitochondrial protease ClpP, and paxalisib, which inhibits PI3K/mTOR, respectively. Results ONC201 and paxalisib combination treatment aimed at inducing metabolic distress led to the design of the first DMG-specific platform trial PNOC022 (NCT05009992). Conclusions Here, we expand on the PNOC022 rationale and discuss various considerations, including liquid biome, microbiome, and genomic biomarkers, quality-of-life endpoints, and novel imaging modalities, such that we offer direction on future clinical trials in DMG.
Collapse
Affiliation(s)
- Sabine Mueller
- Department of Neurology, Neurosurgery and Pediatrics, University of California, San Francisco, California, USA
| | - Cassie Kline
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Andrea Franson
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Michael Prados
- Department of Neurosurgery and Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | - Sebastian M Waszak
- Department of Neurology, University of California, San Francisco, San Francisco, California, USA
- Laboratory of Computational Neuro-Oncology, Swiss Institute for Experimental Cancer Research, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | | - Annette M Molinaro
- Division of Biomedical Statistics and Informatics, Department of Neurosurgery, University of California, San Francisco, San Francisco, California, USA
| | - Carl Koschmann
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | - Javad Nazarian
- Research Center for Genetic Medicine, Children’s National Health System, Washington, District of Columbia, USA
- Brain Tumor Institute, Children’s National Health System, Washington, District of Columbia, USA
- DMG Research Center, Department of Pediatrics, University Children’s Hospital, University of Zurich, Zürich, Switzerland
| |
Collapse
|
7
|
Dimentberg E, Marceau MP, Lachance A, Bergeron-Gravel S, Saikali S, Crevier L, Bourget C, Hawkins C, Jabado N, Giannakouros P, Renzi S, Larouche V. Very Long-term Survivorship in Pediatric DIPG: Case Report and Review of the Literature. J Pediatr Hematol Oncol 2024; 46:211-215. [PMID: 38573000 DOI: 10.1097/mph.0000000000002853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/29/2024] [Indexed: 04/05/2024]
Abstract
Diffuse intrinsic pontine gliomas are lethal tumors with a prognosis generally less than 1 year. Few cases of survivors of 5 years or more have been reported. This case report highlights the journey of a 9.5-year survivor who underwent 3 rounds of focal radiotherapy; she experienced 6 years of progression-free survival following the first round but ultimately succumbed to her disease. An autopsy revealed a favorable IDH1 mutation and the absence of H3K27M. This case reiterates the importance of extensive molecular analyses in diffuse intrinsic pontine gliomas and explores the potential benefit of re-irradiation in patients with positive responses and long periods of remission.
Collapse
Affiliation(s)
| | | | | | | | - Stephan Saikali
- Department of Molecular Biology, Medical Biology, and Pathology
| | | | | | - Cynthia Hawkins
- Department of Pediatrics, Division of Hemato-Oncology, CHU de Québec-Université Laval, Quebec
| | - Nada Jabado
- Department of Laboratory Medicine and Pathobiology - Neuropathology, The Hospital for Sick Children, Toronto
| | - Panagiota Giannakouros
- Department of Pediatrics, Division of Hemato-Oncology, Montreal Children's Hospital, Montreal, Canada
| | - Samuele Renzi
- Department of Pediatrics, Division of Hemato-Oncology, Montreal Children's Hospital, Montreal, Canada
| | - Valérie Larouche
- Department of Pediatrics, Division of Hemato-Oncology, Montreal Children's Hospital, Montreal, Canada
| |
Collapse
|
8
|
Peterson ER, Sajjakulnukit P, Scott AJ, Heaslip C, Andren A, Wilder-Romans K, Zhou W, Palavalasa S, Korimerla N, Lin A, O'Brien A, Kothari A, Zhao Z, Zhang L, Morgan MA, Venneti S, Koschmann C, Jabado N, Lyssiotis CA, Castro MG, Wahl DR. Purine salvage promotes treatment resistance in H3K27M-mutant diffuse midline glioma. Cancer Metab 2024; 12:11. [PMID: 38594734 PMCID: PMC11003124 DOI: 10.1186/s40170-024-00341-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 03/21/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND Diffuse midline gliomas (DMG), including diffuse intrinsic pontine gliomas (DIPGs), are a fatal form of brain cancer. These tumors often carry a driver mutation on histone H3 converting lysine 27 to methionine (H3K27M). DMG-H3K27M are characterized by altered metabolism and resistance to standard of care radiation (RT) but how the H3K27M mediates the metabolic response to radiation and consequent treatment resistance is uncertain. METHODS We performed metabolomics on irradiated and untreated H3K27M isogenic DMG cell lines and observed an H3K27M-specific enrichment for purine synthesis pathways. We profiled the expression of purine synthesis enzymes in publicly available patient data and our models, quantified purine synthesis using stable isotope tracing, and characterized the in vitro and in vivo response to de novo and salvage purine synthesis inhibition in combination with RT. RESULTS DMG-H3K27M cells activate purine metabolism in an H3K27M-specific fashion. In the absence of genotoxic treatment, H3K27M-expressing cells have higher relative activity of de novo synthesis and apparent lower activity of purine salvage demonstrated via stable isotope tracing of key metabolites in purine synthesis and by lower expression of hypoxanthine-guanine phosphoribosyltransferase (HGPRT), the rate-limiting enzyme of purine salvage into IMP and GMP. Inhibition of de novo guanylate synthesis radiosensitized DMG-H3K27M cells in vitro and in vivo. Irradiated H3K27M cells upregulated HGPRT expression and hypoxanthine-derived guanylate salvage but maintained high levels of guanine-derived salvage. Exogenous guanine supplementation decreased radiosensitization in cells treated with combination RT and de novo purine synthesis inhibition. Silencing HGPRT combined with RT markedly suppressed DMG-H3K27M tumor growth in vivo. CONCLUSIONS Our results indicate that DMG-H3K27M cells rely on highly active purine synthesis, both from the de novo and salvage synthesis pathways. However, highly active salvage of free purine bases into mature guanylates can bypass inhibition of the de novo synthetic pathway. We conclude that inhibiting purine salvage may be a promising strategy to overcome treatment resistance in DMG-H3K27M tumors.
Collapse
Affiliation(s)
- Erik R Peterson
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Peter Sajjakulnukit
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Andrew J Scott
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Caleb Heaslip
- Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA
| | - Anthony Andren
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Kari Wilder-Romans
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Weihua Zhou
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Sravya Palavalasa
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Navyateja Korimerla
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Angelica Lin
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Alexandra O'Brien
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Ayesha Kothari
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Zitong Zhao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Li Zhang
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Meredith A Morgan
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Sriram Venneti
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Carl Koschmann
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Nada Jabado
- Department of Pediatrics, McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Costas A Lyssiotis
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Maria G Castro
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, USA
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Daniel R Wahl
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA.
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.
- Medical Science Unit I, 1301 Catherine Street, Rm 4433, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
9
|
Brown EJ, Balaguer-Lluna L, Cribbs AP, Philpott M, Campo L, Browne M, Wong JF, Oppermann U, Carcaboso ÁM, Bullock AN, Farnie G. PRMT5 inhibition shows in vitro efficacy against H3K27M-altered diffuse midline glioma, but does not extend survival in vivo. Sci Rep 2024; 14:328. [PMID: 38172189 PMCID: PMC10764357 DOI: 10.1038/s41598-023-48652-x] [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: 04/05/2023] [Accepted: 11/29/2023] [Indexed: 01/05/2024] Open
Abstract
H3K27-altered Diffuse Midline Glioma (DMG) is a universally fatal paediatric brainstem tumour. The prevalent driver mutation H3K27M creates a unique epigenetic landscape that may also establish therapeutic vulnerabilities to epigenetic inhibitors. However, while HDAC, EZH2 and BET inhibitors have proven somewhat effective in pre-clinical models, none have translated into clinical benefit due to either poor blood-brain barrier penetration, lack of efficacy or toxicity. Thus, there remains an urgent need for new DMG treatments. Here, we performed wider screening of an epigenetic inhibitor library and identified inhibitors of protein arginine methyltransferases (PRMTs) among the top hits reducing DMG cell viability. Two of the most effective inhibitors, LLY-283 and GSK591, were targeted against PRMT5 using distinct binding mechanisms and reduced the viability of a subset of DMG cells expressing wild-type TP53 and mutant ACVR1. RNA-sequencing and phenotypic analyses revealed that LLY-283 could reduce the viability, clonogenicity and invasion of DMG cells in vitro, representing three clinically important phenotypes, but failed to prolong survival in an orthotopic xenograft model. Together, these data show the challenges of DMG treatment and highlight PRMT5 inhibitors for consideration in future studies of combination treatments.
Collapse
Affiliation(s)
- Elizabeth J Brown
- Nuffield Department of Medicine, Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Leire Balaguer-Lluna
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Adam P Cribbs
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, National Institute of Health Research Oxford Biomedical Research Unit (BRU), University of Oxford, Oxford, UK
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, UK
| | - Martin Philpott
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, National Institute of Health Research Oxford Biomedical Research Unit (BRU), University of Oxford, Oxford, UK
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, UK
| | - Leticia Campo
- Department of Oncology, Experimental Cancer Medicine Centre, University of Oxford, Oxford, UK
| | - Molly Browne
- Department of Oncology, Experimental Cancer Medicine Centre, University of Oxford, Oxford, UK
| | - Jong Fu Wong
- Nuffield Department of Medicine, Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Udo Oppermann
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, National Institute of Health Research Oxford Biomedical Research Unit (BRU), University of Oxford, Oxford, UK
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, UK
| | - Ángel M Carcaboso
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Alex N Bullock
- Nuffield Department of Medicine, Centre for Medicines Discovery, University of Oxford, Oxford, UK.
| | - Gillian Farnie
- Nuffield Department of Medicine, Centre for Medicines Discovery, University of Oxford, Oxford, UK.
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, UK.
- Cancer Research Horizons, The Francis Crick Institute, London, UK.
| |
Collapse
|
10
|
Spencer D, Bonner ER, Tor-Díez C, Liu X, Bougher K, Prasad R, Gordish-Dressman H, Eze A, Packer RJ, Nazarian J, Linguraru MG, Bornhorst M. Tumor volume features predict survival outcomes for patients diagnosed with diffuse intrinsic pontine glioma. Neurooncol Adv 2024; 6:vdae151. [PMID: 39434924 PMCID: PMC11492488 DOI: 10.1093/noajnl/vdae151] [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] [Indexed: 10/23/2024] Open
Abstract
Background Diffuse intrinsic pontine glioma (DIPG) is a fatal childhood central nervous system tumor. Diagnosis and monitoring of tumor response to therapy is based on magnetic resonance imaging (MRI). MRI-based analyses of tumor volume and appearance may aid in the prediction of patient overall survival (OS). Methods Contrast-enhanced T1- and FLAIR/T2-weighted MR images were retrospectively collected from children with classical DIPG diagnosed by imaging (n = 43 patients). MRI features were evaluated at diagnosis (n = 43 patients) and post-radiation (n = 40 patients) to determine OS outcome predictors. Features included 3D tumor volume (Twv), contrast-enhancing tumor core volume (Tc), Tc relative to Twv (TC/Twv), and Twv relative to whole brain volume. Support vector machine (SVM) learning was used to identify feature combinations that predicted OS outcome (defined as OS shorter or longer than 12 months from diagnosis). Results Features associated with poor OS outcome included the presence of contrast-enhancing tumor at diagnosis, >15% Tc/Twv post-radiation therapy (RT), and >20% ∆Tc/Twv post-RT. Consistently, SVM learning identified Tc/Twv at diagnosis (prediction accuracy of 74%) and ∆Tc/Twv at <2 months post-RT (accuracy = 75%) as primary features of poor survival. Conclusions This study demonstrates that tumor imaging features at diagnosis and within 4 months of RT can predict differential OS outcomes in DIPG. These findings provide a framework for incorporating tumor volume-based predictive analyses into the clinical setting, with the potential for treatment customization based on tumor risk characteristics and future applications of machine-learning-based analysis.
Collapse
Affiliation(s)
- D’Andre Spencer
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, District of Columbia, USA
- Institute for Clinical and Translational Science, University of California, Irvine, California, USA
| | - Erin R Bonner
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, District of Columbia, USA
| | - Carlos Tor-Díez
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, District of Columbia, USA
| | - Xinyang Liu
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, District of Columbia, USA
| | - Kristen Bougher
- School of Medicine and Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Rachna Prasad
- Department of Oncology, University Children’s Hospital Zürich, Zürich, Switzerland
| | - Heather Gordish-Dressman
- Department of Biostatistics, Children’s National Hospital, Washington, District of Columbia, USA
| | - Augustine Eze
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, District of Columbia, USA
| | - Roger J Packer
- Brain Tumor Institute, Children’s National Hospital, Washington, District of Columbia, USA
| | - Javad Nazarian
- Brain Tumor Institute, Children’s National Hospital, Washington, District of Columbia, USA
- School of Medicine and Health Sciences, The George Washington University, Washington, District of Columbia, USA
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, District of Columbia, USA
| | - Marius George Linguraru
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, District of Columbia, USA
| | - Miriam Bornhorst
- Stanley Manne Children’s Research Institute at Lurie Children’s, Chicago, Illinois, USA
- Department of Hematology, Oncology, Neuro-oncology and Stem Cell Transplant, Ann & Robert H. Lurie Children’s Hospital of Chicago, Illinois, USA
- Brain Tumor Institute, Children’s National Hospital, Washington, District of Columbia, USA
- School of Medicine and Health Sciences, The George Washington University, Washington, District of Columbia, USA
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, District of Columbia, USA
| |
Collapse
|
11
|
Gong X, Kuang S, Deng D, Wu J, Zhang L, Liu C. Differences in survival prognosticators between children and adults with H3K27M-mutant diffuse midline glioma. CNS Neurosci Ther 2023; 29:3863-3875. [PMID: 37311690 PMCID: PMC10651973 DOI: 10.1111/cns.14307] [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: 04/05/2023] [Revised: 05/28/2023] [Accepted: 05/30/2023] [Indexed: 06/15/2023] Open
Abstract
AIMS H3K27M-mutant diffuse midline glioma (DMG) is a rare and aggressive central nervous system tumor. The biological behavior, clinicopathological characteristics, and prognostic factors of DMG have not yet been completely uncovered, especially in adult patients. This study aims to investigate the clinicopathological characteristics and identify prognostic factors of H3K27M-mutant DMG in pediatric and adult patients, respectively. METHODS A total of 171 patients with H3K27M-mutant DMG were included in the study. The clinicopathological characteristics of the patients were analyzed and stratified based on age. The Cox proportional hazard model was used to determine the independent prognostic factors in pediatric and adult subgroups. RESULTS The median overall survival (OS) for the entire cohort was 9.0 months. Significant differences were found in some clinicopathological characteristics between children and adults. The median OS was also significantly different between the pediatric and adult subgroups, with 7.1 months for children and 12.3 months for adults (p < 0.001). In the overall population, the multivariate analysis identified adult patients, single lesion, concurrent chemoradiotherapy/radiotherapy, and intact ATRX expression as independent favorable prognostic factors. In the age-stratified subgroups, the prognostic factors varied between children and adults, with intact ATRX expression and single lesion being independent favorable prognostic factors in adults, while infratentorial localization was significantly associated with worse prognosis in children. CONCLUSIONS The differences in clinicopathological features and prognostic factors between pediatric and adult patients with H3K27M-mutant DMG suggest the need for further clinical and molecular stratification based on age.
Collapse
Affiliation(s)
- Xuan Gong
- Departments of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
| | - Shuwen Kuang
- Departments of OncologyXiangya Hospital, Central South UniversityChangshaChina
| | - Dongfeng Deng
- Departments of OncologyXiangya Hospital, Central South UniversityChangshaChina
| | - Jun Wu
- Departments of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
| | - Longbo Zhang
- Departments of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaChina
| | - Chao Liu
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
- Departments of OncologyXiangya Hospital, Central South UniversityChangshaChina
| |
Collapse
|
12
|
Tosi U, Souweidane M. Fifty years of DIPG: looking at the future with hope. Childs Nerv Syst 2023; 39:2675-2686. [PMID: 37382660 DOI: 10.1007/s00381-023-06037-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 06/17/2023] [Indexed: 06/30/2023]
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a primary brainstem tumor of childhood that carries a dismal prognosis, with median survival of less than 1 year. Because of the brain stem location and pattern of growth within the pons, Dr. Harvey Cushing, the father of modern neurosurgery, urged surgical abandonment. Such a dismal prognosis remained unchanged for decades, coupled with a lack of understanding of tumor biology and an unchanging therapeutic panorama. Beyond palliative external beam radiation therapy, no therapeutic approach has been widely accepted. In the last one to two decades, however, increased tissue availability, an improving understanding of biology, genetics, and epigenetics have led to the development of novel therapeutic targets. In parallel with this biological revolution, new methods intended to enhance drug delivery into the brain stem are contributing to a surge of exciting experimental therapeutic strategies.
Collapse
Affiliation(s)
- Umberto Tosi
- Department of Neurosurgery, Weill Cornell Medicine, 525 E 68th St Box 99, New York, NY, 10021, USA
| | - Mark Souweidane
- Department of Neurosurgery, Weill Cornell Medicine, 525 E 68th St Box 99, New York, NY, 10021, USA.
| |
Collapse
|
13
|
Peterson ER, Sajjakulnukit P, Scott AJ, Heaslip C, Andren A, Wilder-Romans K, Zhou W, Palavalasa S, Korimerla N, Lin A, Obrien A, Kothari A, Zhao Z, Zhang L, Morgan MA, Venneti S, Koschmann C, Jabado N, Lyssiotis CA, Castro MG, Wahl DR. Adaptive rewiring of purine metabolism promotes treatment resistance in H3K27M-mutant diffuse midline glioma. RESEARCH SQUARE 2023:rs.3.rs-3317816. [PMID: 37790517 PMCID: PMC10543500 DOI: 10.21203/rs.3.rs-3317816/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Background Diffuse midline gliomas (DMG), including diffuse intrinsic pontine gliomas (DIPGs), are a fatal form of brain cancer. These tumors often carry a driver mutation on histone H3 converting lysine 27 to methionine (H3K27M). DMG-H3K27M are characterized by altered metabolism and resistance to standard of care radiation (RT), but how the H3K27M mediates the metabolic response to radiation and consequent treatment resistance is uncertain. Methods We performed metabolomics on irradiated and untreated H3K27M isogenic DMG cell lines and observed an H3K27M-specific enrichment for purine synthesis pathways. We profiled the expression of purine synthesis enzymes in publicly available patient data and in our models, quantified purine synthetic flux using stable isotope tracing, and characterized the in vitro and in vivo response to de novo and salvage purine synthesis inhibition in combination with RT. Results DMG-H3K27M cells activate purine metabolism in an H3K27M-specific fashion. In the absence of genotoxic treatment, H3K27M-expressing cells have higher relative activity of de novosynthesis and lower activity of purine salvage due to decreased expression of the purine salvage enzymes. Inhibition of de novo synthesis radiosensitized DMG-H3K27M cells in vitro and in vivo. Irradiated H3K27M cells adaptively upregulate purine salvage enzyme expression and pathway activity. Silencing the rate limiting enzyme in purine salvage, hypoxanthine guanine phosphoribosyl transferase (HGPRT) when combined with radiation markedly suppressed DMG-H3K27M tumor growth in vivo. Conclusions H3K27M expressing cells rely on de novo purine synthesis but adaptively upregulate purine salvage in response to RT. Inhibiting purine salvage may help overcome treatment resistance in DMG-H3K27M tumors.
Collapse
Affiliation(s)
| | | | | | - Caleb Heaslip
- Massachusetts College of Pharmacy and Health Sciences
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Pan C, Zhang M, Xiao X, Kong L, Wu Y, Zhao X, Sun T, Zhang P, Geng Y, Zuo P, Wang Y, Li X, Gu G, Li T, Wu Z, Zhang J, Zhang L. A multimodal imaging-based classification for pediatric diffuse intrinsic pontine gliomas. Neurosurg Rev 2023; 46:151. [PMID: 37358632 DOI: 10.1007/s10143-023-02068-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/31/2023] [Accepted: 06/20/2023] [Indexed: 06/27/2023]
Abstract
OBJECT Pediatric diffuse intrinsic pontine glioma (DIPG) is a radiologically heterogeneous disease entity, here we aim to establish a multimodal imaging-based radiological classification and evaluate the outcome of different treatment strategies under this classification frame. METHODS This retrospective study included 103 children diagnosed with DIPGs between January 2015 and August 2018 in Beijing Tiantan Hospital (Beijing, China). Multimodal radiological characteristics, including conventional magnetic resonance imaging (MRI), diffuse tensor imaging/diffuse tensor tractography (DTI/DTT), and positron emission tomography (PET) were reviewed to construct the classification. The outcome of different treatment strategies was compared in each DIPG subgroup using Kaplan-Meier method (log-rank test) to determine the optimal treatment for specific DIPGs. RESULTS Four radiological DIPG types were identified: Type A ("homocentric", n=13), Type B ("ventral", n=41), Type C ("eccentric", n=37), and Type D ("dorsal", n=12). Their treatment modalities were grouped as observation (43.7%), cytoreductive surgery (CRS) plus radiotherapy (RT) (24.3%), RT alone (11.7%), and CRS alone (20.4%). CRS+RT mainly fell into type C (29.7%), followed by type B1 (21.9%) and type D (50%). Overall, CRS+RT exhibited a potential survival advantage compared to RT alone, which was more pronounced in specific type, but this did not reach statistical significance, due to limited sample size and unbalanced distribution. CONCLUSION We proposed a multimodality imaging-based radiological classification for pediatric DIPG, which was useful for selecting optimal treatment strategies, especially for identifying candidates who may benefit from CRS plus RT. This classification opened a window into image-guided integrated treatment for pediatric DIPG.
Collapse
Affiliation(s)
- Changcun Pan
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070, China
| | - Mingxin Zhang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070, China
| | - Xiong Xiao
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070, China
| | - Lu Kong
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070, China
| | - Yuliang Wu
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070, China
| | - Xiaobin Zhao
- Department of Nuclear Medicine, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070, China
| | - Tao Sun
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070, China
| | - Peng Zhang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070, China
| | - Yibo Geng
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070, China
| | - Pengcheng Zuo
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070, China
| | - Yi Wang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070, China
| | - Xiaoou Li
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070, China
| | - Guocan Gu
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070, China
| | - Tian Li
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070, China
| | - Zhen Wu
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070, China
| | - Junting Zhang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070, China
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing Tian Tan Hospital, Beijing, 100070, China.
| |
Collapse
|
15
|
Knowles T, Huang T, Qi J, An S, Burket N, Cooper S, Nazarian J, Saratsis AM. LIN28B and Let-7 in Diffuse Midline Glioma: A Review. Cancers (Basel) 2023; 15:3241. [PMID: 37370851 DOI: 10.3390/cancers15123241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/12/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023] Open
Abstract
Diffuse midline glioma (DMG) is the most lethal of all childhood cancers. DMGs are driven by histone-tail-mutation-mediated epigenetic dysregulation and partner mutations in genes controlling proliferation and migration. One result of this epigenetic and genetic landscape is the overexpression of LIN28B RNA binding protein. In other systems, LIN28B has been shown to prevent let-7 microRNA biogenesis; however, let-7, when available, faithfully suppresses tumorigenic pathways and induces cellular maturation by preventing the translation of numerous oncogenes. Here, we review the current literature on LIN28A/B and the let-7 family and describe their role in gliomagenesis. Future research is then recommended, with a focus on the mechanisms of LIN28B overexpression and localization in DMG.
Collapse
Affiliation(s)
- Truman Knowles
- W.M. Keck Science Department, Scripps, Pitzer, and Claremont McKenna Colleges, Claremont, CA 91711, USA
| | - Tina Huang
- Department of Neurosurgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jin Qi
- Department of Neurosurgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Shejuan An
- Department of Neurosurgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Noah Burket
- Department of Neurosurgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Scott Cooper
- Department of Neurosurgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Javad Nazarian
- Department of Pediatrics, Children's National Hospital, Washington, DC 20010, USA
- Department of Pediatrics, Zurich Children's Hospital, 8032 Zurich, Switzerland
| | - Amanda M Saratsis
- Department of Neurosurgery, Lutheran General Hospital, Park Ridge, IL 60068, USA
| |
Collapse
|
16
|
Noon A, Galban S. Therapeutic avenues for targeting treatment challenges of diffuse midline gliomas. Neoplasia 2023; 40:100899. [PMID: 37030112 PMCID: PMC10119952 DOI: 10.1016/j.neo.2023.100899] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/24/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023]
Abstract
Diffuse midline glioma (DMG) is the leading cause of brain tumor-related deaths in children. DMG typically presents with variable neurologic symptoms between ages 3 and 10. Currently, radiation remains the standard therapy for DMG to halt progression and reduce tumor bulk to minimize symptoms. However, tumors recur in almost 100% of patients and thus, DMG is still considered an incurable cancer with a median survival of 9-12 months. Surgery is generally contraindicated due to the delicate organization of the brainstem, where DMG is located. Despite extensive research efforts, no chemotherapeutic agents, immune therapies, or molecularly targeted therapies have been approved to provide survival benefit. Furthermore, the efficacy of therapies is limited by poor blood-brain barrier penetration and inherent resistance mechanisms of the tumor. However, novel drug delivery approaches, along with recent advances in molecularly targeted therapies and immunotherapies, have advanced to clinical trials and may provide viable future treatment options for DMG patients. This review seeks to evaluate current therapeutics at the preclinical stage and those that have advanced to clinical trials and to discuss the challenges of drug delivery and inherent resistance to these therapies.
Collapse
Affiliation(s)
- Aleeha Noon
- College of Medicine, California Northstate University, 9700 W Taron Drive, Elk Grove, CA 95757, USA
| | - Stefanie Galban
- Center for Molecular Imaging, The University of Michigan Medical School, BSRB A502, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA; Department of Radiology, The University of Michigan Medical School, BSRB A502, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA; Rogel Cancer Center, The University of Michigan Medical School, 1500 E Medical Center Drive, Ann Arbor, MI 48109, USA.
| |
Collapse
|
17
|
Liu C, Kuang S, Wu L, Cheng Q, Gong X, Wu J, Zhang L. Radiotherapy and radio-sensitization in H3 K27M -mutated diffuse midline gliomas. CNS Neurosci Ther 2023. [PMID: 37157237 DOI: 10.1111/cns.14225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 05/10/2023] Open
Abstract
BACKGROUND H3K27M mutated diffuse midline gliomas (DMGs) are extremely aggressive and the leading cause of cancer-related deaths in pediatric brain tumors with 5-year survival <1%. Radiotherapy is the only established adjuvant treatment of H3K27M DMGs; however, the radio-resistance is commonly observed. METHODS We summarized current understandings of the molecular responses of H3K27M DMGs to radiotherapy and provide crucial insights into current advances in radiosensitivity enhancement. RESULTS Ionizing radiation (IR) can mainly inhibit tumor cell growth by inducing DNA damage regulated by the cell cycle checkpoints and DNA damage repair (DDR) system. In H3K27M DMGs, the aberrant genetic and epigenetic changes, stemness genotype, and epithelial-mesenchymal transition (EMT) disrupt the cell cycle checkpoints and DDR system by altering the associated regulatory signaling pathways, which leads to the development of radio-resistance. CONCLUSIONS The advances in mechanisms of radio-resistance in H3K27M DMGs promote the potential targets to enhance the sensitivity to radiotherapy.
Collapse
Affiliation(s)
- Chao Liu
- Departments of Oncology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Shuwen Kuang
- Departments of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Quan Cheng
- Departments of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xuan Gong
- Departments of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jun Wu
- Departments of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Longbo Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Departments of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Departments of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
18
|
Lyu Y, Guo Y, Okeoma CM, Yan Z, Hu N, Li Z, Zhou S, Zhao X, Li J, Wang X. Engineered extracellular vesicles (EVs): Promising diagnostic/therapeutic tools for pediatric high-grade glioma. Biomed Pharmacother 2023; 163:114630. [PMID: 37094548 DOI: 10.1016/j.biopha.2023.114630] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/29/2023] [Accepted: 03/29/2023] [Indexed: 04/26/2023] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a highly malignant brain tumor that mainly occurs in children with extremely low overall survival. Traditional therapeutic strategies, such as surgical resection and chemotherapy, are not feasible mostly due to the special location and highly diffused features. Radiotherapy turns out to be the standard treatment method but with limited benefits of overall survival. A broad search for novel and targeted therapies is in the progress of both preclinical investigations and clinical trials. Extracellular vesicles (EVs) emerged as a promising diagnostic and therapeutic candidate due to their distinct biocompatibility, excellent cargo-loading-delivery capacity, high biological barrier penetration efficiency, and ease of modification. The utilization of EVs in various diseases as biomarker diagnoses or therapeutic agents is revolutionizing modern medical research and practice. In this review, we will briefly talk about the research development of DIPG, and present a detailed description of EVs in medical applications, with a discussion on the application of engineered peptides on EVs. The possibility of applying EVs as a diagnostic tool and drug delivery system in DIPG is also discussed.
Collapse
Affiliation(s)
- Yuan Lyu
- Medical Research Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Henan Joint International Laboratory of Glioma Metabolism and Microenvironment Research, Henan Provincial Department of Science and Technology, Zhengzhou, Henan 450052, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yupei Guo
- Henan Joint International Laboratory of Glioma Metabolism and Microenvironment Research, Henan Provincial Department of Science and Technology, Zhengzhou, Henan 450052, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Chioma M Okeoma
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY 10595-1524, USA
| | - Zhaoyue Yan
- Department of Neurosurgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Nan Hu
- Henan Joint International Laboratory of Glioma Metabolism and Microenvironment Research, Henan Provincial Department of Science and Technology, Zhengzhou, Henan 450052, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zian Li
- Henan Joint International Laboratory of Glioma Metabolism and Microenvironment Research, Henan Provincial Department of Science and Technology, Zhengzhou, Henan 450052, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Shaolong Zhou
- Henan Joint International Laboratory of Glioma Metabolism and Microenvironment Research, Henan Provincial Department of Science and Technology, Zhengzhou, Henan 450052, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xin Zhao
- Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Junqi Li
- Medical Research Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Henan Joint International Laboratory of Glioma Metabolism and Microenvironment Research, Henan Provincial Department of Science and Technology, Zhengzhou, Henan 450052, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Xinjun Wang
- Henan Joint International Laboratory of Glioma Metabolism and Microenvironment Research, Henan Provincial Department of Science and Technology, Zhengzhou, Henan 450052, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Neurosurgery, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| |
Collapse
|
19
|
Hart E', Bianco J, Bruin MAC, Derieppe M, Besse HC, Berkhout K, Kie LACJ, Su Y, Hoving EW, Huitema ADR, Ries MG, van Vuurden DG. Radiosensitisation by olaparib through focused ultrasound delivery in a diffuse midline glioma model. J Control Release 2023; 357:287-298. [PMID: 37019285 DOI: 10.1016/j.jconrel.2023.03.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/21/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND AND PURPOSE Diffuse midline glioma H3K27-altered (DMG) is an aggressive, inoperable, predominantly paediatric brain tumour. Treatment strategies are limited, resulting in a median survival of only 11 months. Currently, radiotherapy (RT), often combined with temozolomide, is considered the standard of care but remains palliative, highlighting the urgency for new therapies. Radiosensitisation by olaparib, an inhibitor of PARP1 and subsequently PAR-synthesis, is a promising treatment option. We assessed whether PARP1 inhibition enhances radiosensitivity in vitro and in vivo following focused ultrasound mediated blood-brain barrier opening (FUS-BBBO). METHODS Effects of PARP1 inhibition were evaluated in vitro using viability, clonogenic, and neurosphere assays. In vivo olaparib extravasation and pharmacokinetic profiling following FUS-BBBO was measured by LC-MS/MS. Survival benefit of FUS-BBBO combined with olaparib and RT was assessed using a patient-derived xenograft (PDX) DMG mouse model. RESULTS Treatment with olaparib in combination with radiation delayed tumour cell proliferation in vitro through the reduction of PAR. Prolonged exposure of low olaparib concentration was more efficient in delaying cell growth than short exposure of high concentration. FUS-BBBO increased olaparib bioavailability in the pons by 5.36-fold without observable adverse effects. A Cmax of 54.09 μM in blood and 1.39 μM in the pontine region was achieved following administration of 100 mg/kg olaparib. Although RT combined with FUS-BBBO mediated olaparib extravasation delayed local tumour growth, survival benefits were not observed in an in vivo DMG PDX model. CONCLUSIONS Olaparib effectively radiosensitises DMG cells in vitro and reduces primary tumour growth in vivo when combined with RT. Further studies are needed to investigate the therapeutic benefit of olaparib in suitable preclinical PDX models.
Collapse
Affiliation(s)
- E 't Hart
- Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
| | - J Bianco
- Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands.
| | - M A C Bruin
- Department of Pharmacy and Pharmacology, the Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands
| | - M Derieppe
- Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
| | - H C Besse
- Center for Imaging Sciences, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - K Berkhout
- Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
| | - L A Chin Joe Kie
- Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
| | - Y Su
- Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
| | - E W Hoving
- Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
| | - A D R Huitema
- Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands; Department of Pharmacy and Pharmacology, the Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands; Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - M G Ries
- Center for Imaging Sciences, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - D G van Vuurden
- Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
| |
Collapse
|
20
|
Fiz F, Bottoni G, Ugolini M, Righi S, Cirone A, Garganese MC, Verrico A, Rossi A, Milanaccio C, Ramaglia A, Mastronuzzi A, Abate ME, Cacchione A, Gandolfo C, Colafati GS, Garrè ML, Morana G, Piccardo A. Diagnostic and Dosimetry Features of [ 64Cu]CuCl 2 in High-Grade Paediatric Infiltrative Gliomas. Mol Imaging Biol 2023; 25:391-400. [PMID: 36042116 DOI: 10.1007/s11307-022-01769-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/10/2022] [Accepted: 08/18/2022] [Indexed: 03/12/2023]
Abstract
PURPOSE OF THE REPORT Paediatric diffuse high-grade gliomas (PDHGG) are rare central nervous system neoplasms lacking effective therapeutic options. Molecular imaging of tumour metabolism might identify novel diagnostic/therapeutic targets. In this study, we evaluated the distribution and the dosimetry aspects of [64Cu]CuCl2 in PDHGG subjects, as copper is a key element in cellular metabolism whose turnover may be increased in tumour cells. MATERIAL AND METHODS Paediatric patients with PDHGG were prospectively recruited. [64Cu]CuCl2 PET/CT was performed 1 h after tracer injection; if the scan was positive, it was repeated 24 and 72 h later. Lesion standardised uptake value (SUV) and target-to-background ratio (TBR) were calculated. Tumour and organ dosimetry were computed using the MIRD algorithm. Each patient underwent an MRI scan, including FLAIR, T2-weighted and post-contrast T1-weighted imaging. RESULTS Ten patients were enrolled (median age 9, range 6-16 years, 6 females). Diagnoses were diffuse midline gliomas (n = 8, 5 of which with H3K27 alterations) and diffuse hemispheric gliomas (n = 2). Six patients had visible tracer uptake (SUV: 1.0 ± 0.6 TBR: 5 ± 3.1). [64Cu]CuCl2 accumulation was always concordant with MRI contrast enhancement and was higher in the presence of radiological signs of necrosis. SUV and TBR progressively increased on the 24- and 72-h acquisitions (p < 0.05 and p < 0.01, respectively). The liver and the abdominal organs received the highest non-target dose. CONCLUSIONS [64Cu]CuCl2 is a well-tolerated radiotracer with reasonably favourable dosimetric properties, showing selective uptake in tumour areas with visible contrast enhancement and necrosis, thus suggesting that blood-brain barrier damage is a pre-requisite for its distribution to the intracranial structures. Moreover, tracer uptake showed an accumulating trend over time. These characteristics could deserve further analysis, to determine whether this radiopharmaceutical might have a possible therapeutic role as well.
Collapse
Affiliation(s)
- Francesco Fiz
- Department of Nuclear Medicine, E.O. Ospedali Galliera, Galliera Hospital, Mura delle Cappuccine 14, 16128, Genoa, Italy.
| | - Gianluca Bottoni
- Department of Nuclear Medicine, E.O. Ospedali Galliera, Galliera Hospital, Mura delle Cappuccine 14, 16128, Genoa, Italy
| | - Martina Ugolini
- Department of Nuclear Medicine, E.O. Ospedali Galliera, Galliera Hospital, Mura delle Cappuccine 14, 16128, Genoa, Italy
| | - Sergio Righi
- Medical Physics Department, E.O. Galliera Hospital, Genoa, Italy
| | - Alessio Cirone
- Medical Physics Department, E.O. Galliera Hospital, Genoa, Italy
| | - Maria Carmen Garganese
- Nuclear Medicine Unit/Imaging Department, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Antonio Verrico
- Neuro-Oncology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Andrea Rossi
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy.,Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | | | - Antonia Ramaglia
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Angela Mastronuzzi
- Neuro-Oncology Unit, Department of Paediatric Haematology/Oncology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
| | | | - Antonella Cacchione
- Neuro-Oncology Unit, Department of Paediatric Haematology/Oncology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
| | - Carlo Gandolfo
- Imaging Department, Neuroradiology Unit, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | | | | | - Giovanni Morana
- Department of Neurosciences, University of Turin, Turin, Italy
| | - Arnoldo Piccardo
- Department of Nuclear Medicine, E.O. Ospedali Galliera, Galliera Hospital, Mura delle Cappuccine 14, 16128, Genoa, Italy
| |
Collapse
|
21
|
Kim HJ, Suh CO. Radiotherapy for Diffuse Intrinsic Pontine Glioma: Insufficient but Indispensable. Brain Tumor Res Treat 2023; 11:79-85. [PMID: 37151149 PMCID: PMC10172015 DOI: 10.14791/btrt.2022.0041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/28/2022] [Accepted: 01/02/2023] [Indexed: 05/09/2023] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) account for 10%-20% of all central nervous system tumors in children and are the leading cause of death in children with brain tumors. Although many clinical trials have been conducted over the past decades, the survival outcome has remained unchanged. Over 90% of children die within 2 years of the diagnosis, and radiotherapy remains the standard treatment to date. To improve the prognosis, hyperfractionated and hypofractionated radiotherapy and/or addition of radiosensitizers have been investigated. However, none of the radiotherapy approaches have shown a survival benefit, and the overall survival of patients with DIPG is approximately 11 months. Here, we comprehensively review the management of DIPG with focus on radiotherapy.
Collapse
Affiliation(s)
- Hyun Ju Kim
- Department of Radiation Oncology, Gachon University Gil Hospital, Gachon University College of Medicine, Incheon, Korea
| | - Chang-Ok Suh
- Department of Radiation Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea.
| |
Collapse
|
22
|
Park J, Chung C. Epigenetic and Metabolic Changes in Diffuse Intrinsic Pontine Glioma. Brain Tumor Res Treat 2023; 11:86-93. [PMID: 37151150 PMCID: PMC10172016 DOI: 10.14791/btrt.2023.0011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/07/2023] [Accepted: 04/13/2023] [Indexed: 05/09/2023] Open
Abstract
Diffuse midline glioma (DMG), hitherto known as diffuse intrinsic pontine glioma (DIPG), is a rare and aggressive form of brain cancer that primarily affects children. Although the exact cause of DMG/DIPG is not known, a large proportion of DMG/DIPG tumors harbor mutations in the gene encoding the histone H3 protein, specifically the H3K27M mutation. This mutation decreases the level of H3K27me3, a histone modification that plays a vital role in regulating gene expression through epigenetic regulation. The mutation also alters the function of polycomb repressive complex 2 (PRC2), thereby preventing the repression of genes associated with cancer development. The decrease in H3K27me3 caused by the histone H3 mutation is accompanied by an increase in the level of H3K27ac, a post-translational modification related to active transcription. Dysregulation of histone modification markedly affects gene expression, contributing to cancer development and progression by promoting uncontrolled cell proliferation, tumor growth, and metabolism. DMG/DIPG alters the metabolism of methionine and the tricarboxylic acid cycle, as well as glucose and glutamine uptake. The role of epigenetic and metabolic changes in the development of DMG/DIPG has been studied extensively, and understanding these changes is critical to developing therapies targeting these pathways. Studies are currently underway to identify new therapeutic targets for DMG/DIPG, which may lead to the development of effective treatments for this devastating disease.
Collapse
Affiliation(s)
- Jiyoon Park
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Korea
- New Biology Research Center (NBRC), Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Korea
| | - Chan Chung
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Korea
- New Biology Research Center (NBRC), Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Korea.
| |
Collapse
|
23
|
Parsels LA, Wahl DR, Koschmann C, Morgan MA, Zhang Q. Developing H3K27M mutant selective radiosensitization strategies in diffuse intrinsic pontine glioma. Neoplasia 2023; 37:100881. [PMID: 36724689 PMCID: PMC9918797 DOI: 10.1016/j.neo.2023.100881] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/13/2023] [Accepted: 01/23/2023] [Indexed: 01/31/2023]
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a rare but highly lethal pediatric and adolescent tumor located in the pons of the brainstem. DIPGs harbor unique and specific pathological and molecular alterations, such as the hallmark lysine 27-to-methionine (H3K27M) mutation in histone H3, which lead to global changes in the epigenetic landscape and drive tumorigenesis. While fractionated radiotherapy, the current standard of care, improves symptoms and delays tumor progression, DIPGs inevitably recur, and despite extensive efforts chemotherapy-driven radiosensitization strategies have failed to improve survival. Advances in our understanding of the role of epigenetics in the cellular response to radiation-induced DNA damage, however, offer new opportunities to develop combinational therapeutic strategies selective for DIPGs expressing H3K27M. In this review, we provide an overview of preclinical studies that explore potential radiosensitization strategies targeting the unique epigenetic landscape of H3K27M mutant DIPG. We further discuss opportunities to selectively radiosensitize DIPG through strategic inhibition of the radiation-induced DNA damage response. Finally, we discuss the potential for using radiation to induce anti-tumor immune responses that may be potentiated in DIPG by radiosensitizing-therapeutic strategies.
Collapse
Affiliation(s)
- Leslie A Parsels
- Department of Radiation Oncology, Rogel Cancer Center, University of Michigan Medical School, 1301 Catherine Street, Ann Arbor, MI, 48109, USA
| | - Daniel R Wahl
- Department of Radiation Oncology, Rogel Cancer Center, University of Michigan Medical School, 1301 Catherine Street, Ann Arbor, MI, 48109, USA
| | - Carl Koschmann
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Meredith A Morgan
- Department of Radiation Oncology, Rogel Cancer Center, University of Michigan Medical School, 1301 Catherine Street, Ann Arbor, MI, 48109, USA.
| | - Qiang Zhang
- Department of Radiation Oncology, Rogel Cancer Center, University of Michigan Medical School, 1301 Catherine Street, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
24
|
Towards Standardisation of a Diffuse Midline Glioma Patient-Derived Xenograft Mouse Model Based on Suspension Matrices for Preclinical Research. Biomedicines 2023; 11:biomedicines11020527. [PMID: 36831063 PMCID: PMC9952880 DOI: 10.3390/biomedicines11020527] [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: 12/15/2022] [Revised: 01/31/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Diffuse midline glioma (DMG) is an aggressive brain tumour with high mortality and limited clinical therapeutic options. Although in vitro research has shown the effectiveness of medication, successful translation to the clinic remains elusive. A literature search highlighted the high variability and lack of standardisation in protocols applied for establishing the commonly used HSJD-DIPG-007 patient-derived xenograft (PDX) model, based on animal host, injection location, number of cells inoculated, volume, and suspension matrices. This study evaluated the HSJD-DIPG-007 PDX model with respect to its ability to mimic human disease progression for therapeutic testing in vivo. The mice received intracranial injections of HSJD-DIPG-007 cells suspended in either PBS or Matrigel. Survival, tumour growth, and metastases were assessed to evaluate differences in the suspension matrix used. After cell implantation, no severe side effects were observed. Additionally, no differences were detected in terms of survival or tumour growth between the two suspension groups. We observed delayed metastases in the Matrigel group, with a significant difference compared to mice with PBS-suspended cells. In conclusion, using Matrigel as a suspension matrix is a reliable method for establishing a DMG PDX mouse model, with delayed metastases formation and is a step forward to obtaining a standardised in vivo PDX model.
Collapse
|
25
|
Zhang P, Duan Y, Gu G, Qu L, Xiao D, Xi T, Pan C, Liu Y, Zhang L. Clinical, pathological, and radiological features of 80 pediatric diffuse intrinsic pontine gliomas: A single-institute study. Front Oncol 2023; 13:1007393. [PMID: 36824137 PMCID: PMC9941347 DOI: 10.3389/fonc.2023.1007393] [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: 07/30/2022] [Accepted: 01/16/2023] [Indexed: 02/10/2023] Open
Abstract
Objective Diffuse intrinsic pontine gliomas (DIPGs) are rare but devastating diseases. This retrospective cross-sectional study aimed to investigate the clinical, radiological, and pathological features of DIPGs. Materials and methods The clinical data of 80 pediatric DIPGs under clinical treatment in Beijing Tiantan Hospital from July 2013 to July 2019 were retrospectively collected and studied. A follow-up evaluation was performed. Results This study included 48 men and 32 women. The most common symptoms were cranial nerve palsy (50.0%, 40/80 patients) and limb weakness (41.2%, 33/80 patients). Among the 80 patients, 24 cases were clinically diagnosed, 56 cases were pathologically verified, and 45 cases were tested for H3K27 alteration status, with 34 H3K27 alteration cases confirmed. Radiological results indicated that enhancement was common (65.0%, 52/80 patients). Cho/Cr was of predictive value for H3K27 alteration status (P = 0.012, cutoff value = 2.38, AUC = 0.801). Open cranial surgery followed by further chemotherapy and radiotherapy was beneficial for patients' overall survival. Cox regression analysis indicated H3K27 alteration to be the independent prognostic influencing factor for DIPGs in this series (P = 0.002). Conclusion DIPGs displayed a wide spectrum of clinical and imaging features. Surgery-suitable patients could benefit from postoperative comprehensive therapy for a better overall survival. H3K27 alteration was the independent prognostic influencing factor for DIPGs.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China,Beijing Neurosurgical Institute, Beijing, China
| | - Yunyun Duan
- China National Clinical Research Center for Neurological Diseases, Beijing, China,Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guocan Gu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Liying Qu
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Dan Xiao
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Tianshu Xi
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Changcun Pan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China,Beijing Neurosurgical Institute, Beijing, China
| | - Ya’ou Liu
- China National Clinical Research Center for Neurological Diseases, Beijing, China,Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China,*Correspondence: Ya’ou Liu, ; Liwei Zhang,
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China,Beijing Neurosurgical Institute, Beijing, China,*Correspondence: Ya’ou Liu, ; Liwei Zhang,
| |
Collapse
|
26
|
Wagner MW, Namdar K, Napoleone M, Hainc N, Amirabadi A, Fonseca A, Laughlin S, Shroff MM, Bouffet E, Hawkins C, Khalvati F, Bartels U, Ertl-Wagner BB. Radiomic Features Based on MRI Predict Progression-Free Survival in Pediatric Diffuse Midline Glioma/Diffuse Intrinsic Pontine Glioma. Can Assoc Radiol J 2023; 74:119-126. [PMID: 35768942 DOI: 10.1177/08465371221109921] [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: 01/11/2023] Open
Abstract
Purpose: Biopsy-based assessment of H3 K27 M status helps in predicting survival, but biopsy is usually limited to unusual presentations and clinical trials. We aimed to evaluate whether radiomics can serve as prognostic marker to stratify diffuse intrinsic pontine glioma (DIPG) subsets. Methods: In this retrospective study, diagnostic brain MRIs of children with DIPG were analyzed. Radiomic features were extracted from tumor segmentations and data were split into training/testing sets (80:20). A conditional survival forest model was applied to predict progression-free survival (PFS) using training data. The trained model was validated on the test data, and concordances were calculated for PFS. Experiments were repeated 100 times using randomized versions of the respective percentage of the training/test data. Results: A total of 89 patients were identified (48 females, 53.9%). Median age at time of diagnosis was 6.64 years (range: 1-16.9 years) and median PFS was 8 months (range: 1-84 months). Molecular data were available for 26 patients (29.2%) (1 wild type, 3 K27M-H3.1, 22 K27M-H3.3). Radiomic features of FLAIR and nonenhanced T1-weighted sequences were predictive of PFS. The best FLAIR radiomics model yielded a concordance of .87 [95% CI: .86-.88] at 4 months PFS. The best T1-weighted radiomics model yielded a concordance of .82 [95% CI: .8-.84] at 4 months PFS. The best combined FLAIR + T1-weighted radiomics model yielded a concordance of .74 [95% CI: .71-.77] at 3 months PFS. The predominant predictive radiomic feature matrix was gray-level size-zone. Conclusion: MRI-based radiomics may predict progression-free survival in pediatric diffuse midline glioma/diffuse intrinsic pontine glioma.
Collapse
Affiliation(s)
- Matthias W Wagner
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada.,Department of Medical Imaging, 7938University of Toronto, Canada
| | - Khashayar Namdar
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada.,Department of Medical Imaging, 7938University of Toronto, Canada
| | - Marc Napoleone
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Nicolin Hainc
- Nicolin Hainc:Department of Neuroradiology, Clinical Neuroscience Center, 7979University Hospital Zurich,University of Zurich, Switzerland
| | - Afsaneh Amirabadi
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Adriana Fonseca
- Department of Neurooncology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Suzanne Laughlin
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada.,Department of Medical Imaging, 7938University of Toronto, Canada
| | - Manohar M Shroff
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada.,Department of Medical Imaging, 7938University of Toronto, Canada
| | - Eric Bouffet
- Department of Neurooncology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Cynthia Hawkins
- Department of Paediatric Laboratory Medicine, Division of Pathology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Farzad Khalvati
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Ute Bartels
- Department of Neurooncology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Birgit B Ertl-Wagner
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada.,Department of Medical Imaging, 7938University of Toronto, Canada
| |
Collapse
|
27
|
11C-methionine PET imaging characteristics in children with diffuse intrinsic pontine gliomas and relationship to survival and H3 K27M mutation status. Eur J Nucl Med Mol Imaging 2023; 50:1709-1719. [PMID: 36697961 DOI: 10.1007/s00259-022-06105-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/30/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE This study aimed to describe 11C-methionine (11C-MET) PET imaging characteristics in patients with paediatric diffuse intrinsic pontine glioma (DIPG) and correlate them with survival and H3 K27M mutation status. METHODS We retrospectively analysed 98 children newly diagnosed with DIPG who underwent 11C-MET PET. PET imaging characteristics evaluated included uptake intensity, uniformity, metabolic tumour volume (MTV), and total lesion methionine uptake (TLMU). The maximum, mean, and peak of the tumour-to-background ratio (TBR), calculated as the corresponding standardised uptake values (SUV) divided by the mean reference value, were also recorded. The associations between the PET imaging characteristics and clinical outcomes in terms of progression-free survival (PFS) and overall survival (OS) and H3 K27M mutation status were assessed, respectively. RESULTS In univariate analysis, imaging characteristics significantly associated with shorter PFS and OS included a higher uniformity grade, higher TBRs, larger MTV, and higher TLMU. In multivariate analysis, larger MTV at diagnosis, shorter symptom duration, and no treatment were significantly correlated with shorter PFS and OS. The PET imaging features were not correlated with H3 K27M mutation status. CONCLUSION Although several imaging features were significantly associated with PFS and OS, only MTV, indicating the size of the active tumour, was identified as a strong independent prognostic factor.
Collapse
|
28
|
Kim HJ, Lee JH, Kim Y, Lim DH, Park SH, Ahn SD, Kim IA, Im JH, Chung JW, Kim JY, Kim IH, Yoon HI, Suh CO. Suggestions for Escaping the Dark Ages for Pediatric Diffuse Intrinsic Pontine Glioma Treated with Radiotherapy: Analysis of Prognostic Factors from the National Multicenter Study. Cancer Res Treat 2023; 55:41-49. [PMID: 35255651 PMCID: PMC9873330 DOI: 10.4143/crt.2021.1514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/02/2022] [Indexed: 02/04/2023] Open
Abstract
PURPOSE This multicenter retrospective study aimed to investigate clinical, radiologic, and treatment-related factors affecting survival in patients with newly diagnosed diffuse intrinsic pontine glioma (DIPG) treated with radiotherapy. MATERIALS AND METHODS Patients aged <30 years who underwent radiotherapy as an initial treatment for DIPG between 2000 and 2018 were included; patients who did not undergo magnetic resonance imaging at diagnosis and those with pathologically diagnosed grade I glioma were excluded. We examined medical records of 162 patients collected from 10 participating centers in Korea. The patients' clinical, radiological, molecular, and histopathologic characteristics, and treatment responses were evaluated to identify the prognosticators for DIPG and estimate survival outcomes. RESULTS The median follow-up period was 10.8 months (interquartile range, 7.5 to 18.1). The 1- and 2-year overall survival (OS) rates were 53.5% and 19.0%, respectively, with a median OS of 13.1 months. Long-term survival rate (≥ 2 years) was 16.7%, and median OS was 43.6 months. Age (< 10 years), poor performance status, treatment before 2010, and post-radiotherapy necrosis were independently associated with poor OS in multivariate analysis. In patients with increased post-radiotherapy necrosis, the median OS estimates were 13.3 months and 11.4 months with and without bevacizumab, respectively (p=0.138). CONCLUSION Therapeutic strategy for DIPG has remained unchanged over time, and the associated prognosis remains poor. Our findings suggest that appropriate efforts are needed to reduce the occurrence of post-radiotherapy necrosis. Further well-designed clinical trials are recommended to improve the poor prognosis observed in DIPG patients.
Collapse
Affiliation(s)
- Hyun Ju Kim
- Department of Radiation Oncology, Gachon University Gil Hospital, Incheon,
Korea
| | - Joo Ho Lee
- Department of Radiation Oncology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul,
Korea
| | - Youngkyong Kim
- Department of Radiation Oncology, Kyung Hee University Medical Center, Kyung Hee University School of Medicine, Seoul,
Korea
| | - Do Hoon Lim
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul,
Korea
| | - Shin-Hyung Park
- Department of Radiation Oncology, School of Medicine, Kyungpook National University, Daegu,
Korea
| | - Seung Do Ahn
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - In Ah Kim
- Department of Radiation Oncology, Seoul National University Bundang Hospital, Seongnam,
Korea
| | - Jung Ho Im
- Department of Radiation Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam,
Korea
| | - Jae Wook Chung
- Department of Radiation Oncology, Chonnam National University Medical School, Gwangju,
Korea
| | - Joo-Young Kim
- Proton Therapy Center, National Cancer Center, Goyang,
Korea
| | - Il Han Kim
- Department of Radiation Oncology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul,
Korea
| | - Hong In Yoon
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul,
Korea
| | - Chang-Ok Suh
- Department of Radiation Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam,
Korea
| |
Collapse
|
29
|
Zhao JP, Liu XJ, Lin HZ, Cui CX, Yue YJ, Gao S, Xu HZ. MRI comparative study of diffuse midline glioma, H3 K27-altered and glioma in the midline without H3 K27-altered. BMC Neurol 2022; 22:498. [PMID: 36550486 PMCID: PMC9773507 DOI: 10.1186/s12883-022-03026-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
PURPOSE The MRI features of Diffuse midline glioma, H3 K27-altered and glioma in the midline without H3 K27-altered were compared and analyzed, and the changes in the apparent diffusion coefficient (ADC) of the two groups were quantitatively analyzed. METHODS The MRI images of 35 patients with Diffuse midline gliomas, H3 K27-altered and gliomas in the midline without H3 K27-altered were analyzed retrospectively. The location, edge, signal, peritumoral edema and enhancement characteristics of the lesions were observed, and the changes in ADC values were analyzed. RESULTS In the H3 K27-altered group, 85.7% (12/14) of the tumors were located in the thalamus and brainstem compared with 28.6% (6/21) in the no H3 K27-altered group. In the H3 K27-altered group, for tumors only located in the midline area, only 14.3% (1/7) had irregular shapes and unclear boundaries, while for tumors also invaded the extramidline tissues 85.7% (6/7) had irregular shapes and unclear boundaries.The"basilar artery wrapped sign" was found in 6 patients with tumors located in the pons in the H3 K27-altered group, but none in the no H3 K27-altered group had this sign. In the H3 K27-altered group, only 14.3% (1/7) of the tumors confined to the midline area had small cystic degeneration and necrosis, while for tumors also invaded the extramidline tissues, 100% (7/7) of the tumors had cystic degeneration and necrosis, and the cystic degeneration and necrosis only located in the extramidline region of the tumor in 6 cases.A total of 78.6% (11/14) of tumors in the H3 K27-altered group showed mild to moderate enhancement, while 47.6% (10/21) of tumors in the no H3 K27-altered group showed mild to moderate enhancement. The average peritumoral edema index was 1.13 in the H3 K27-altered group and 1.75 in the no H3 K27-altered group. The average ADC value of tumor in the H3 K27-altered group was 7.83 × 10- 4 mm2/s, and the ratio to normal brain tissue was 0.844, while the values in the no H3 K27-altered group were 13.5 × 10- 4 mm2/s and 1.75, respectively. CONCLUSION Compared with gliomas in the midline without H3 K27-altered, The MRI findings and ADC value of Diffuse midline gliomas, H3K27-altered have some characteristics, which can help improve the diagnosis and differential diagnosis.
Collapse
Affiliation(s)
- Ji-ping Zhao
- grid.412521.10000 0004 1769 1119Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xue-jun Liu
- grid.412521.10000 0004 1769 1119Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hao-zhi Lin
- grid.412521.10000 0004 1769 1119Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chun-xiao Cui
- grid.412521.10000 0004 1769 1119Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ying-jie Yue
- grid.412521.10000 0004 1769 1119Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Song Gao
- grid.412521.10000 0004 1769 1119Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hong-zhang Xu
- grid.477864.eDepartment of Radiology, Shidao People’s Hospital, Rongcheng, China
| |
Collapse
|
30
|
Developing a Predictive Grading Model for Children with Gliomas Based on Diffusion Kurtosis Imaging Metrics: Accuracy and Clinical Correlations with Patient Survival. Cancers (Basel) 2022; 14:cancers14194778. [PMID: 36230701 PMCID: PMC9563289 DOI: 10.3390/cancers14194778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/19/2022] [Accepted: 09/24/2022] [Indexed: 11/20/2022] Open
Abstract
Purpose: To develop a predictive grading model based on diffusion kurtosis imaging (DKI) metrics in children affected by gliomas, and to investigate the clinical impact of the predictive model by correlating with overall survival and progression-free survival. Materials and methods: 59 patients with a histological diagnosis of glioma were retrospectively studied (33 M, 26 F, median age 7.2 years). Patients were studied on a 3T scanner with a standardized MR protocol, including conventional and DKI sequences. Mean kurtosis (MK), axial kurtosis (AK), radial kurtosis (RK), fractional anisotropy (FA), and apparent diffusion coefficient (ADC) maps were obtained. Whole tumour volumes (VOIs) were segmented semi-automatically. Mean DKI values were calculated for each metric. The quantitative values from DKI-derived metrics were used to develop a predictive grading model to develop a probability prediction of a high-grade glioma (pHGG). Three models were tested: DTI-based, DKI-based, and combined (DTI and DKI). The grading accuracy of the resulting probabilities was tested with a receiver operating characteristics (ROC) analysis for each model. In order to account for dataset imbalances between pLGG and pHGG, we applied a random synthetic minority oversampling technique (SMOTE) analysis. Lastly, the most accurate model predictions were correlated with progression-free survival (PFS) and overall survival (OS) using the Kaplan−Meier method. Results: The cohort included 46 patients with pLGG and 13 patients with pHGG. The developed model predictions yielded an AUC of 0.859 (95%CI: 0.752−0.966) for the DTI model, of 0.939 (95%CI: 0.879−1) for the DKI model, and of 0.946 (95%CI: 0.890−1) for the combined model, including input from both DTI and DKI metrics, which resulted in the most accurate model. Sample estimation with the random SMOTE analysis yielded an AUC of 0.98 on the testing set. Model predictions from the combined model were significantly correlated with PFS (25.2 months for pHGG vs. 40.0 months for pLGG, p < 0.001) and OS (28.9 months for pHGG vs. 44.9 months for pLGG, p < 0.001). Conclusions: a DKI-based predictive model was highly accurate for pediatric glioma grading. The combined model, derived from both DTI and DKI metrics, proved that DKI-based model predictions of tumour grade were significantly correlated with progression-free survival and overall survival.
Collapse
|
31
|
Sun K, Xu M, Fei X, Wang H, Xu L, Xu R, Xu M. Prediction of Cancer-Specific Survival of Brainstem Glioma in Children Based on Risk Stratification Model. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:3436631. [PMID: 35912147 PMCID: PMC9328996 DOI: 10.1155/2022/3436631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/26/2022] [Accepted: 06/23/2022] [Indexed: 11/18/2022]
Abstract
Objective To develop and authenticate a risk stratification framework and nomogram for ascertaining cancer-specific survival (CSS) among the pediatric brainstem gliomas. Methods For patients less than 12 years, according to Surveillance, Epidemiology, and End Results (SEER), information from 1998 to 2016 is found in their databases. The survival outcomes, treatments, and demographic clinicopathologic conditions are scrutinized per the database validation, and training cohorts are divided and validated using multivariate Cox regression analysis. A nomogram was designed, and predominantly, the risk stratification conceptualization engaged selected tenets according to the multivariate analysis. The model's authenticity was substantiated through C-index measure and calibration curves. Results There are 806 pediatric concerns of histologically concluded brainstem glioma in the research. According to multivariate analysis, age, grade, radiotherapy, and race (with P value < 0.05) depicted independent prognostic variations of the pediatric gliomas. The nomogram's C-index was approximately 0.75 and an accompanied predictive capability for CSS. Conclusion The nomogram constructed in this glioma's context is the primary predictor of using risk stratification. A combination of nomograms with the risk stratification mechanism assists clinicians in monitoring high-risk individuals and engage targeted accessory treatment.
Collapse
Affiliation(s)
- Kai Sun
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, China
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Mingwei Xu
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Xiaowei Fei
- Department of Neurosurgery, The First Affiliated Hospital of The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Hao Wang
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Lunshan Xu
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Ruxiang Xu
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Minhui Xu
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| |
Collapse
|
32
|
Bartlett AL, Lane A, Chaney B, Escorza NY, Black K, Cochrane A, Minturn J, Bartels U, Warren K, Hansford J, Ziegler D, Diez B, Goldman S, Packer R, Kieran M, DeWire-Schottmiller M, Erker C, Monje-Deisseroth M, Wagner L, Koschmann C, Dorris K, Shih CS, Hassall T, Samson Y, Fisher P, Wang SS, Tsui K, Sevlever G, Zhu X, Dexheimer P, Asher A, Fuller C, Drissi R, Jones B, Leach J, Fouladi M. Characteristics of children ≤36 months of age with DIPG: A report from the international DIPG registry. Neuro Oncol 2022; 24:2190-2199. [PMID: 35552452 PMCID: PMC9713498 DOI: 10.1093/neuonc/noac123] [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: 12/31/2022] Open
Abstract
BACKGROUND Children ≤36 months with diffuse intrinsic pontine glioma (DIPG) have increased long-term survival (LTS, overall survival (OS) ≥24 months). Understanding distinguishing characteristics in this population is critical to improving outcomes. METHODS Patients ≤36 months at diagnosis enrolled on the International DIPG Registry (IDIPGR) with central imaging confirmation were included. Presentation, clinical course, imaging, pathology and molecular findings were analyzed. RESULTS Among 1183 patients in IDIPGR, 40 were eligible (median age: 29 months). Median OS was 15 months. Twelve patients (30%) were LTS, 3 (7.5%) very long-term survivors ≥5 years. Among 8 untreated patients, median OS was 2 months. Patients enrolled in the registry but excluded from our study by central radiology review or tissue diagnosis had median OS of 7 months. All but 1 LTS received radiation. Among 32 treated patients, 1-, 2-, 3-, and 5-year OS rates were 68.8%, 31.2%, 15.6% and 12.5%, respectively. LTS had longer duration of presenting symptoms (P = .018). No imaging features were predictive of outcome. Tissue and genomic data were available in 18 (45%) and 10 patients, respectively. Among 9 with known H3K27M status, 6 had a mutation. CONCLUSIONS Children ≤36 months demonstrated significantly more LTS, with an improved median OS of 15 months; 92% of LTS received radiation. Median OS in untreated children was 2 months, compared to 17 months for treated children. LTS had longer duration of symptoms. Excluded patients demonstrated a lower OS, contradicting the hypothesis that children ≤36 months with DIPG show improved outcomes due to misdiagnosis.
Collapse
Affiliation(s)
- Allison L Bartlett
- Corresponding Author: Allison Bartlett, MD, 3333 Burnet Ave, MLC 1107, Cincinnati, OH 45229, USA ()
| | - Adam Lane
- Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Brooklyn Chaney
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Nancy Yanez Escorza
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Katie Black
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Anne Cochrane
- Brain Tumor Center, Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA,University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jane Minturn
- Division of Oncology, Children’s Hospital of Philadelphia and Perelman School of Medicine, Philadelphia, Pennsylvania,USA
| | - Ute Bartels
- Department of Pediatrics, Division of Oncology, University of Toronto and The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kathy Warren
- Department of Pediatric Oncology, Dana Farber Cancer Institute/Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Jordan Hansford
- Children’s Cancer Centre, Royal Children’s Hospital; Murdoch Children’s Research Institute; University of Melbourne, Melbourne, Australia
| | - David Ziegler
- Children’s Cancer Institute Australia, Lowy Cancer Research Centre, UNSW and Kids Cancer Centre, Sydney’s Children Hospital, Randwick, Sydney NSW, Australia,School of Women’s and Children’s Health, University of New South Wales, Sydney, Australia
| | - Blanca Diez
- FLENI (Fundacion para Lucha contra las Enfermedes Neurologicas de Infantes), Buenos Aires, Argentina
| | - Stewart Goldman
- Division of Pediatric Hematology and Oncology, Center for Cancer and Blood Disorders, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois,USA
| | - Roger Packer
- Department of Neurology, Center for Neuroscience and Behavioral Medicine, Children’s National Hospital, Washington, DC, USA
| | - Mark Kieran
- Department of Pediatrics, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Mariko DeWire-Schottmiller
- Brain Tumor Center, Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Craig Erker
- Department of Pediatrics, Dalhousie University and IWK Health Center, Halifax, Nova Scotia, Canada
| | - Michelle Monje-Deisseroth
- Department of Neurology, Neurosurgery, Pediatrics, and Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Lars Wagner
- Division of Pediatric Hematology/Oncology, Kentucky Children’s Hospital, University of Kentucky, Lexington, Kentucky, USA
| | - Carl Koschmann
- Department of Pediatrics, C.S. Mott Children’s Hospital and University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Kathleen Dorris
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Chie-Schin Shih
- Division of Hematology/Oncology, Department of Pediatrics, Indiana University School of Medicine, Riley Hospital for Children at Indiana University Health, Indianapolis, Indiana, USA
| | - Tim Hassall
- Queensland Children’s Hospital, Brisbane, Queensland, Australia
| | - Yvan Samson
- Department of Hematology-Oncology, Université de Montréal and CHU Sainte-Justine, Montréal, Québec, Canada
| | - Paul Fisher
- Department of Neurology, Division of Child Neurology, Stanford University, Palo Alto, California, USA
| | - Stacie S Wang
- Children’s Cancer Centre, Royal Children’s Hospital; Murdoch Children’s Research Institute; University of Melbourne, Melbourne, Australia
| | - Karen Tsui
- Starship Blood and Cancer Centre, Starship Children’s Health, Auckland, New Zealand
| | - Gustavo Sevlever
- FLENI (Fundacion para Lucha contra las Enfermedes Neurologicas de Infantes), Buenos Aires, Argentina
| | - Xiaoting Zhu
- Brain Tumor Center, Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA,Department of Electrical Engineering and Computer Science, University of Cincinnati College of Engineering and Applied Science, Cincinnati, Ohio, USA
| | - Phillip Dexheimer
- Department of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio, USA
| | - Anthony Asher
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Christine Fuller
- Department of Pathology, Upstate Medical University, Syracuse, New York, USA
| | - Rachid Drissi
- Center for Childhood Cancer & Blood Disorders, Nationwide Children’s Hospital, Columbus, Ohio, USA,The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Blaise Jones
- Division of Radiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - James Leach
- Division of Radiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Maryam Fouladi
- The Ohio State University College of Medicine, Columbus, Ohio, USA,Pediatric Neuro-Oncology Program, Nationwide Children’s Hospital, Columbus, Ohio, USA
| |
Collapse
|
33
|
Lazow MA, Fuller C, DeWire M, Lane A, Bandopadhayay P, Bartels U, Bouffet E, Cheng S, Cohen KJ, Cooney TM, Coven SL, Dholaria H, Diez B, Dorris K, El-ayadi M, El-Sheikh A, Fisher PG, Fonseca A, Garcia Lombardi M, Greiner RJ, Goldman S, Gottardo N, Gururangan S, Hansford JR, Hassall T, Hawkins C, Kilburn L, Koschmann C, Leary SE, Ma J, Minturn JE, Monje-Deisseroth M, Packer R, Samson Y, Sandler ES, Sevlever G, Tinkle CL, Tsui K, Wagner LM, Zaghloul M, Ziegler DS, Chaney B, Black K, Asher A, Drissi R, Fouladi M, Jones BV, Leach JL. Accuracy of central neuro-imaging review of DIPG compared with histopathology in the International DIPG Registry. Neuro Oncol 2022; 24:821-833. [PMID: 34668975 PMCID: PMC9071293 DOI: 10.1093/neuonc/noab245] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Diffuse intrinsic pontine glioma (DIPG) remains a clinico-radiologic diagnosis without routine tissue acquisition. Reliable imaging distinction between DIPG and other pontine tumors with potentially more favorable prognoses and treatment considerations is essential. METHODS Cases submitted to the International DIPG registry (IDIPGR) with histopathologic and/or radiologic data were analyzed. Central imaging review was performed on diagnostic brain MRIs (if available) by two neuro-radiologists. Imaging features suggestive of alternative diagnoses included nonpontine origin, <50% pontine involvement, focally exophytic morphology, sharply defined margins, and/or marked diffusion restriction throughout. RESULTS Among 286 patients with pathology from biopsy and/or autopsy, 23 (8%) had histologic diagnoses inconsistent with DIPG, most commonly nondiffuse low-grade gliomas and embryonal tumors. Among 569 patients with centrally-reviewed diagnostic MRIs, 40 (7%) were classified as non-DIPG, alternative diagnosis suspected. The combined analysis included 151 patients with both histopathology and centrally-reviewed MRI. Of 77 patients with imaging classified as characteristic of DIPG, 76 (99%) had histopathologic diagnoses consistent with DIPG (infiltrating grade II-IV gliomas). Of 57 patients classified as likely DIPG with some unusual imaging features, 55 (96%) had histopathologic diagnoses consistent with DIPG. Of 17 patients with imaging features suggestive of an alternative diagnosis, eight (47%) had histopathologic diagnoses inconsistent with DIPG (remaining patients were excluded due to nonpontine tumor origin). Association between central neuro-imaging review impression and histopathology was significant (p < 0.001), and central neuro-imaging impression was prognostic of overall survival. CONCLUSIONS The accuracy and important role of central neuro-imaging review in confirming the diagnosis of DIPG is demonstrated.
Collapse
Affiliation(s)
- Margot A Lazow
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Pediatric Neuro-Oncology Program, Nationwide Children’s Hospital, Columbus, Ohio, USA
- The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Christine Fuller
- Department of Pathology, Upstate Medical University, Syracuse, New York, USA
| | - Mariko DeWire
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Adam Lane
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | | | - Ute Bartels
- Division of Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Eric Bouffet
- Division of Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sylvia Cheng
- Division of Pediatric Hematology/Oncology/BMT, British Columbia Children’s Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kenneth J Cohen
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, USA
| | - Tabitha M Cooney
- Dana Farber Cancer Institute, Harvard Cancer Center, Boston, Massachusetts, USA
| | - Scott L Coven
- Division of Oncology, Riley Hospital for Children, Indianapolis, Indiana, USA
| | - Hetal Dholaria
- Department of Oncology, Perth Children’s Hospital, Nedlands, Australia
| | - Blanca Diez
- Department of Oncology and Pathology, Fundacion para la lucha de las enfermedades neurologicas de la infancia FLENI, Buenos Aires, Argentina
| | - Kathleen Dorris
- Center for Cancer and Blood Disorders, Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Moatasem El-ayadi
- National Cancer Institute, Cairo University and Children’s Cancer Hospital Egypt, Cairo, Egypt
| | - Ayman El-Sheikh
- Division of Oncology, Dayton Children’s Hospital, Dayton, Ohio, USA
| | - Paul G Fisher
- Department of Neurology, Stanford University, Stanford, California, USA
| | - Adriana Fonseca
- Division of Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Robert J Greiner
- Division of Oncology, Penn State Health Children’s Hospital, Hershey, Pennsylvania, USA
| | - Stewart Goldman
- Department of Pediatrics, Phoenix Children’s Hospital, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA
| | - Nicholas Gottardo
- Department of Oncology, Perth Children’s Hospital, Nedlands, Australia
| | | | - Jordan R Hansford
- Children’s Cancer Centre, Royal Children’s Hospital Murdoch Children’s Research Institute University of Melbourne, Melbourne, Victoria, Australia
| | - Tim Hassall
- Division of Oncology, Queensland Children’s Hospital, South Brisbane, Australia
| | - Cynthia Hawkins
- Division of Pathology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lindsay Kilburn
- Division of Oncology, Children’s National Medical Center, Washinton, District of Columbia, USA
| | - Carl Koschmann
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | - Sarah E Leary
- Cancer and Blood Disorders Center, Seattle Children’s Hospital, Seattle, Washington, USA
| | - Jie Ma
- Division of Oncology, Xinhua Hospital, Shanghai, China
| | - Jane E Minturn
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Michelle Monje-Deisseroth
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Roger Packer
- Division of Oncology, Children’s National Medical Center, Washinton, District of Columbia, USA
| | - Yvan Samson
- Division of Oncology, CHU Saint Justine, Montreal, Quebec, Canada
| | - Eric S Sandler
- Division of Oncology, Nemours Children’s Health System, Wilmington, Delaware, USA
| | - Gustavo Sevlever
- Department of Oncology and Pathology, Fundacion para la lucha de las enfermedades neurologicas de la infancia FLENI, Buenos Aires, Argentina
| | - Christopher L Tinkle
- Division of Radiation Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Karen Tsui
- Division of Oncology, Starship Children’s Hospital, Auckland, New Zealand
| | - Lars M Wagner
- Division of Pediatric Hematology/Oncology, University of Kentucky, Lexington, Kentucky, USA
| | - Mohamed Zaghloul
- National Cancer Institute, Cairo University and Children’s Cancer Hospital Egypt, Cairo, Egypt
| | - David S Ziegler
- School of Women’s and Children’s Health and Children’s Cancer Institute, University of New South Wales, Sydney, Australia
| | - Brooklyn Chaney
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Katie Black
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Anthony Asher
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Rachid Drissi
- The Ohio State University College of Medicine, Columbus, Ohio, USA
- Center for Childhood Cancer & Blood Disorders, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Maryam Fouladi
- Pediatric Neuro-Oncology Program, Nationwide Children’s Hospital, Columbus, Ohio, USA
- The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Blaise V Jones
- Department of Radiology and Medical Imaging, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - James L Leach
- Department of Radiology and Medical Imaging, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
34
|
Persson ML, Douglas AM, Alvaro F, Faridi P, Larsen MR, Alonso MM, Vitanza NA, Dun MD. The intrinsic and microenvironmental features of diffuse midline glioma; implications for the development of effective immunotherapeutic treatment strategies. Neuro Oncol 2022; 24:1408-1422. [PMID: 35481923 PMCID: PMC9435509 DOI: 10.1093/neuonc/noac117] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Diffuse midline glioma (DMG), including those of the brainstem (diffuse intrinsic pontine glioma), are pediatric tumors of the central nervous system (CNS). Recognized as the most lethal of all childhood cancers, palliative radiotherapy remains the only proven treatment option, however, even for those that respond, survival is only temporarily extended. DMG harbor an immunologically “cold” tumor microenvironment (TME) with few infiltrating immune cells. The mechanisms underpinning the cold TME are not well understood. Low expression levels of immune checkpoint proteins, including PD-1, PD-L1, and CTLA-4, are recurring features of DMG and likely contribute to the lack of response to immune checkpoint inhibitors (ICIs). The unique epigenetic signatures (including stem cell-like methylation patterns), a low tumor mutational burden, and recurring somatic mutations (H3K27M, TP53, ACVR1, MYC, and PIK3CA), possibly play a role in the reduced efficacy of traditional immunotherapies. Therefore, to circumvent the lack of efficacy thus far seen for the use of ICIs, adoptive cell transfer (including chimeric antigen receptor T cells) and the use of oncolytic viruses, are currently being evaluated for the treatment of DMG. It remains an absolute imperative that we improve our understanding of DMG’s intrinsic and TME features if patients are to realize the potential benefits offered by these sophisticated treatments. Herein, we summarize the limitations of immunotherapeutic approaches, highlight the emerging safety and clinical efficacy shown for sophisticated cell-based therapies, as well as the evolving knowledge underpinning the DMG-immune axis, to guide the development of immunotherapies that we hope will improve outcomes.
Collapse
Affiliation(s)
- Mika L Persson
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia.,Precision Medicine Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Alicia M Douglas
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia.,Precision Medicine Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Frank Alvaro
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia.,Precision Medicine Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,John Hunter Children's Hospital, New Lambton Heights, NSW, Australia
| | - Pouya Faridi
- Department of Medicine, School of Clinical Sciences, Monash University, Melbourne, VIC, Australia
| | - Martin R Larsen
- Department of Molecular Biology and Biochemistry, Protein Research Group, University of Southern Denmark, Odense, Denmark
| | - Marta M Alonso
- Department of Pediatrics, University Hospital of Navarra, Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for Applied Medical Research (CIMA), Pamplona, Spain
| | - Nicholas A Vitanza
- The Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA.,Division of Pediatric Hematology, Oncology, Bone Marrow Transplant, and Cellular Therapy, Department of Pediatrics, Seattle Children's Hospital, Seattle, WA, USA
| | - Matthew D Dun
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia.,Precision Medicine Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
35
|
Bronk JK, Hou P, Amsbaugh MJ, Khatua S, Mahajan A, Ketonen L, McGovern SL. Sequential Diffusion Tensor Imaging and Magnetic Resonance Spectroscopy in Patients Undergoing Reirradiation for Progressive Diffuse Intrinsic Pontine Glioma. Adv Radiat Oncol 2022; 7:100847. [PMID: 35071836 PMCID: PMC8763636 DOI: 10.1016/j.adro.2021.100847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 10/31/2021] [Indexed: 11/29/2022] Open
Abstract
Purpose Diffusion tensor imaging for evaluation of white matter tracts is used with magnetic resonance spectroscopy (MRS) to improve management of diffuse intrinsic pontine glioma (DIPG). Changes in the apparent diffusion coefficient (ADC), fractional anisotropy (FA), and tumor metabolite ratios have been reported after initial radiation for DIPG, but these markers have not been studied sequentially in patients undergoing reirradiation for progressive DIPG. Here, we report a case series of 4 patients who received reirradiation for progressive DIPG on a prospective clinical trial in which we evaluated quantitative changes in FA, ADC, and tumor metabolites and qualitative changes in white matter tracts. Methods and Materials The median reirradiation dose was 25.2 Gy (24-30.8 Gy). Fiber tracking was performed using standard tractography analysis. The FA and ADC values for the corticospinal and medial lemniscus tracts were calculated before and after reirradiation. Multivoxel MRS was performed. Findings were correlated with clinical features and conventional MRI of tumors. Results All patients had an initial response to reirradiation as shown by a decrease in tumor size. In general, FA increased with disease response and decreased with progression, whereas ADC decreased with disease response and increased with progression. At second progression, the FA fold change relative to values during disease response decreased in both patients with available imaging at second progression. Visualization of tracts demonstrated robust reconstitution of previously disrupted paths during tumor response; conversely, there was increased fiber tract disruption and infiltration during tumor progression. The MRS analysis revealed a decrease in choline:creatinine and choline:N-acetylaspartate ratios during tumor response and increase during progression. Conclusions Distinct changes in white matter tracts and tumor metabolism were observed in patients with DIPG undergoing reirradiation on a prospective clinical trial. Changes related to tumor response and progression were observed after 24 to 30.8 Gy reirradiation.
Collapse
Affiliation(s)
- Julianna K. Bronk
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ping Hou
- Departments of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark J. Amsbaugh
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Soumen Khatua
- Departments of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anita Mahajan
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Leena Ketonen
- Departments of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Susan L. McGovern
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Corresponding author: Susan L. McGovern, MD, PhD
| |
Collapse
|
36
|
Wu C, Zheng H, Li J, Zhang Y, Duan S, Li Y, Wang D. MRI-based radiomics signature and clinical factor for predicting H3K27M mutation in pediatric high-grade gliomas located in the midline of the brain. Eur Radiol 2022; 32:1813-1822. [PMID: 34655310 DOI: 10.1007/s00330-021-08234-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/11/2021] [Accepted: 07/26/2021] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To develop a nomogram based on MRI radiomics and clinical features for preoperatively predicting H3K27M mutation in pediatric high-grade gliomas (pHGGs) with a midline location of the brain. METHODS The institutional database was reviewed to identify patients with pHGGs with a midline location of the brain who underwent tumor biopsy with preoperative MRI scans between June 2016 and June 2021. A total of 107 patients with pHGGs, including 79 patients with H3K27M mutation, were consecutively included and randomly divided into training and test sets. Radiomics features were extracted from fluid-attenuated inversion recovery (FLAIR), diffusion-weighted (DW) and post-contrast T1-weighted images, and apparent diffusion coefficient (ADC) maps. The minimum redundancy maximum relevance (MRMR) and least absolute shrinkage and selection operator (LASSO) logistic regression were performed for radiomics signature construction. Clinical and radiological features were analyzed to select clinical predictors. A nomogram was then developed by incorporating the radiomics signature and selected clinical predictors. RESULTS Nine radiomics features were selected to construct the radiomics signature, which showed a favorable discriminatory ability in training and test sets with an area under the curve (AUC) of 0.95 and 0.92, respectively. Ring enhancement was identified as an independent clinical predictor (p < 0.01). The nomogram, constructed with radiomics signature and ring enhancement, showed good calibration and discrimination in training and testing sets (AUC: 0.95 and 0.90 respectively). CONCLUSIONS The nomogram which combined radiomics signature and ring enhancement had a satisfactory ability to predict H3K27M mutation in pHGGs with a midline of the brain. KEY POINTS • Conventional MRI features were not powerful enough to predict H3K27M mutation status in pediatric high-grade gliomas (pHGGs) with a midline location of the brain. • An MRI-based radiomics signature showed satisfactory ability to predict H3K27M mutation status of pHGGs located in the midline of the brain. • Associating the radiomics signature with clinical factors improved predictive performance.
Collapse
Affiliation(s)
- Chenqing Wu
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Hui Zheng
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Jinning Li
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yuzhen Zhang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Shaofeng Duan
- GE Healthcare, Pudong New Town, No.1 Huatuo Road, Shanghai, 210000, China
| | - Yuhua Li
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Dengbin Wang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
37
|
Ikeda K, Kolakshyapati M, Takayasu T, Amatya VJ, Takano M, Yonezawa U, Taguchi A, Onishi S, Takeshima Y, Sugiyama K, Yamasaki F. Diffusion-weighted imaging-gadolinium enhancement mismatch sign in diffuse midline glioma. Eur J Radiol 2022; 147:110103. [DOI: 10.1016/j.ejrad.2021.110103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 12/04/2021] [Accepted: 12/08/2021] [Indexed: 11/03/2022]
|
38
|
Evans M, Gill R, Bull KS. Does a Bevacizumab-based regime have a role in the treatment of children with diffuse intrinsic pontine glioma? A systematic review. Neurooncol Adv 2022; 4:vdac100. [PMID: 35821674 PMCID: PMC9270727 DOI: 10.1093/noajnl/vdac100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background There are no effective treatments for diffuse intrinsic pontine glioma (DIPG); median survival is 11.2 months. Bevacizumab has the potential to improve quality of life (QOL) and survival in DIPG but has never been evaluated systematically. The aim of this review was to assess Bevacizumab’s role in the treatment of DIPG. Methods MEDLINE, EMBASE, Scopus, and Web of Science were searched for relevant studies using terms developed from alternatives for Bevacizumab and DIPG. One reviewer screened titles and abstracts, then two reviewers screened full texts. Data were extracted into tables and quality assessed using methodological index for non-randomized studies and JBI tools. Results Searching revealed 1001 papers; after deduplication 851 remained. After screening of titles and abstracts, then 28 full texts, 11 studies were included. Four studies reported a median overall survival longer than historical data, however, two found no significant impact of Bevacizumab. Five studies reported a radiological response in a proportion of participants and two reported no response. Three studies, evaluating clinical response, reported improvement in a proportion of patients. Three studies, evaluating QOL, reported stability or improvement. Four studies, evaluating steroid use, reported reductions in the proportion of patients receiving steroids. In radiation necrosis treatment, Bevacizumab led to clinical improvement in 6/12 patients in 2 studies and permitted a reduction in steroid use in most patients. Conclusions Insufficient evidence means the role of Bevacizumab in the treatment of DIPG is unclear. However, Bevacizumab may be beneficial to some patients. The review highlights the need for further research in this area.
Collapse
Affiliation(s)
- Mia Evans
- Faculty of medicine, University of Southampton , Southampton , UK
| | - Ria Gill
- Faculty of medicine, University of Southampton , Southampton , UK
| | - Kim S Bull
- Clinical and Experimental Sciences, University of Southampton , Southampton , UK
| |
Collapse
|
39
|
Jia H, Zhang P, Gu G, Li T, Jiang Z, Wu Z, Wang L, Zhang J, Duan Y, Liu Y, Yang F, Qin S, Zhang L. Brainstem tumors may increase the impairment of behavioral emotional cognition in children. J Neurooncol 2022; 160:423-432. [PMID: 36333568 PMCID: PMC9722802 DOI: 10.1007/s11060-022-04161-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE It remains unclear as to whether patients with brainstem tumor experience complex neuropsychiatric problems. In this cohort study, we specifically investigated behavioral, emotional and cognitive symptoms in pediatric patients with brainstem glioma and healthy individuals. METHODS A total of 146 patients with pediatric brainstem tumors (aged 4-18 years old) and 46 age-matched healthy children were recruited to assess their behaviors and emotions examined by the Child Behavior Checklist. A variety of clinical factors were also analyzed. RESULTS There were significant differences in most behavioral and emotional symptoms between pediatric patients and healthy subjects. Moreover, patients with pons tumors exhibited significantly higher scores than patients with medulla oblongata tumors (p = 0.012), particularly in concerning the syndrome categories of Withdrawn (p = 0.043), Anxious/depressed symptoms (p = 0.046), Thought Problems (p = 0.004), Attention deficits (p = 0.008), Externalizing problems (p = 0.013), and Aggressive behavior (p = 0.004). A tumor body located in the pontine (p = 0.01, OR = 4.5, 95% CI = 1.4-14.059) or DIPG in the midbrain (p = 0.002, OR = 3.818, 95% CI = 1.629-8.948) appears to act as a risk factor that is associated with more problems in patients with neuropsychiatric symptoms. CONCLUSIONS Pediatric patients with brainstem tumors exhibit severe behavioral and emotional problems. Tumor invades the pontine and midbrain act a risk factor with more problems. It suggests that structural and functional abnormalities in the brainstem will cause prolonged behavioral problems and emotional-cognitive dysfunctions in young children.
Collapse
Affiliation(s)
- Heyuan Jia
- School of Instrumentation and Optoelectronic Engineering, Beihang University, No.37. BeiHang University XueYuan Road, HaiDian District, Beijing, 100083, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, BeiHang University XueYuan Road, HaiDian District, BeiJing, 100083, China
| | - Peng Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119. the West Southern 4Th Ring Road, Fengtai District, Beijing, 100073, China
| | - Guocan Gu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119. the West Southern 4Th Ring Road, Fengtai District, Beijing, 100073, China
| | - Tian Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119. the West Southern 4Th Ring Road, Fengtai District, Beijing, 100073, China
| | - Zhuang Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119. the West Southern 4Th Ring Road, Fengtai District, Beijing, 100073, China
| | - Zhen Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119. the West Southern 4Th Ring Road, Fengtai District, Beijing, 100073, China
| | - Liang Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119. the West Southern 4Th Ring Road, Fengtai District, Beijing, 100073, China
| | - Junting Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119. the West Southern 4Th Ring Road, Fengtai District, Beijing, 100073, China
| | - Yunyun Duan
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, No. 119. the West Southern 4Th Ring Road, Fengtai District, Beijing, 100073, China
| | - Yaou Liu
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, No. 119. the West Southern 4Th Ring Road, Fengtai District, Beijing, 100073, China
| | - Feng Yang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, No. 119. the West Southern 4Th Ring Road, Fengtai District, Beijing, 100073, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100070, China.
| | - Shaozheng Qin
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, No. 19, XinJieKouWai St., HaiDian District, Beijing, 100875, China.
| | - Liwei Zhang
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, BeiHang University XueYuan Road, HaiDian District, BeiJing, 100083, China.
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119. the West Southern 4Th Ring Road, Fengtai District, Beijing, 100073, China.
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, No. 119. the West Southern 4Th Ring Road, Fengtai District, Beijing, 100073, China.
| |
Collapse
|
40
|
Hohm A, Karremann M, Gielen GH, Pietsch T, Warmuth-Metz M, Vandergrift LA, Bison B, Stock A, Hoffmann M, Pham M, Kramm CM, Nowak J. Magnetic Resonance Imaging Characteristics of Molecular Subgroups in Pediatric H3 K27M Mutant Diffuse Midline Glioma. Clin Neuroradiol 2021; 32:249-258. [PMID: 34919158 PMCID: PMC8894220 DOI: 10.1007/s00062-021-01120-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/28/2021] [Indexed: 11/28/2022]
Abstract
Purpose Recent research identified histone H3 K27M mutations to be associated with a dismal prognosis in pediatric diffuse midline glioma (pDMG); however, data on detailed MRI characteristics with respect to H3 K27 mutation status and molecular subgroups (H3.1 and H3.3 K27M mutations) are limited. Methods Standardized magnetic resonance imaging (MRI) parameters and epidemiologic data of 68 pDMG patients (age <18 years) were retrospectively reviewed and compared in a) H3 K27M mutant versus H3 K27 wildtype (WT) tumors and b) H3.1 versus H3.3 K27M mutant tumors. Results Intracranial gliomas (n = 58) showed heterogeneous phenotypes with isointense to hyperintense signal in T2-weighted images and frequent contrast enhancement. Hemorrhage and necrosis may be present. Comparing H3 K27M mutant to WT tumors, there were significant differences in the following parameters: i) tumor localization (p = 0.001), ii) T2 signal intensity (p = 0.021), and iii) T1 signal homogeneity (p = 0.02). No significant imaging differences were found in any parameter between H3.1 and H3.3 K27M mutant tumors; however, H3.1 mutant tumors occurred at a younger age (p = 0.004). Considering spinal gliomas (n = 10) there were no significant imaging differences between the analyzed molecular groups. Conclusion With this study, we are the first to provide detailed MR imaging data on H3 K27M mutant pDMG with respect to molecular subgroup status in a large patient cohort. Our findings may support diagnosis and future targeted therapeutic trials of pDMG within the framework of the radiogenomics concept. Supplementary Information The online version of this article (10.1007/s00062-021-01120-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Annika Hohm
- Neuroradiological Reference Center for the Pediatric Brain Tumor (HIT) Studies of the German Society of Pediatric Oncology and Hematology, Würzburg University Hospital, Würzburg, Germany
- Department of Neuroradiology, Würzburg University Hospital, Würzburg, Germany
- Current address: Division of Pediatric Stem Cell Transplantation and Immunology, University Children's Medical Clinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Karremann
- Department of Pediatric and Adolescent Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gerrit H Gielen
- Institute of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Torsten Pietsch
- Institute of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Monika Warmuth-Metz
- Neuroradiological Reference Center for the Pediatric Brain Tumor (HIT) Studies of the German Society of Pediatric Oncology and Hematology, Würzburg University Hospital, Würzburg, Germany
- Department of Neuroradiology, Würzburg University Hospital, Würzburg, Germany
| | - Lindsey A Vandergrift
- Departments of Radiology and Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Brigitte Bison
- Neuroradiological Reference Center for the Pediatric Brain Tumor (HIT) Studies of the German Society of Pediatric Oncology and Hematology, Würzburg University Hospital, Würzburg, Germany
- Current address: Neuroradiological Reference Center for the Pediatric Brain Tumor (HIT) Studies of the German Society of Pediatric Oncology and Hematology, Department of Neuroradiology, University Augsburg, Faculty of Medicine, Augsburg, Germany
| | - Annika Stock
- Neuroradiological Reference Center for the Pediatric Brain Tumor (HIT) Studies of the German Society of Pediatric Oncology and Hematology, Würzburg University Hospital, Würzburg, Germany
- Department of Neuroradiology, Würzburg University Hospital, Würzburg, Germany
| | - Marion Hoffmann
- Division of Pediatric Hematology and Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Mirko Pham
- Department of Neuroradiology, Würzburg University Hospital, Würzburg, Germany
| | - Christof M Kramm
- Division of Pediatric Hematology and Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Johannes Nowak
- Neuroradiological Reference Center for the Pediatric Brain Tumor (HIT) Studies of the German Society of Pediatric Oncology and Hematology, Würzburg University Hospital, Würzburg, Germany.
- Department of Neuroradiology, Würzburg University Hospital, Würzburg, Germany.
- SRH Poliklinik Gera GmbH, Radiology Gotha, Gotha, Germany.
| |
Collapse
|
41
|
Seong M, Kim ST, Noh JH, Kim YK, Kim HJ. Radiologic findings and the molecular expression profile of diffuse midline glioma H3 K27M mutant. Acta Radiol 2021; 62:1404-1411. [PMID: 33175579 DOI: 10.1177/0284185120968560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Diffuse midline glioma H3 K27M-mutant (DMG) are reported to show heterogeneous radiologic imaging features in children. We hypothesized that other genetic mutations may contribute to this heterogeneity. PURPOSE To describe the magnetic resonance imaging (MRI) findings of DMG in adult patients and to correlate the imaging findings with the molecular expression profile. MATERIAL AND METHODS Eighteen patients with pathologically proven DMG were enrolled. On preoperative MRI, the following were evaluated: location; size of the lesion; ratio of non-enhancing (NE) and contrast-enhancing (CE) area; presence of cortical invasion and necrotic component; maximum relative cerebral blood volume ratio (rCBV ratio) of NE and CE portions; and minimum apparent diffusion coefficient (ADC) of NE and CE portions, among others. Molecular profiles including ATRX expression and p53 mutation were reviewed to find correlation with imaging features. RESULTS Thalamus was the most commonly involved location, followed by pons and tectum. Five patients showed loss of normal ATRX expression. p53 mutation was positive in 12 patients. 40% of normal ATRX expression patients had cortical involvement and 20% had leptomeningeal seeding; none of the patients with ATRX loss had cortical involvement or leptomeningeal seeding. Patients with normal ATRX expression showed significantly higher rCBV ratio and lower ADC value in the NE area than patients with ATRX loss (P=0.04, 0.016). p53 mutation status did not correlate with any imaging finding. CONCLUSION Cortical invasion, leptomeningeal tumor spread, lower ADC value and higher rCBV ratio in NE areas of DMG may be related to normal expression of ATRX.
Collapse
Affiliation(s)
- Minjung Seong
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sung Tae Kim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jung Hyun Noh
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yi Kyung Kim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyung-Jin Kim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
42
|
El-Khouly FE, Adil SM, Wiese M, Hulleman E, Hendrikse NH, Kaspers GJL, Kramm CM, Veldhuijzen van Zanten SEM, van Vuurden DG. Complementary and alternative medicine in children with diffuse intrinsic pontine glioma-A SIOPE DIPG Network and Registry study. Pediatr Blood Cancer 2021; 68:e29061. [PMID: 33942498 DOI: 10.1002/pbc.29061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/27/2021] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Diffuse intrinsic pontine glioma (DIPG) is a rare and aggressive childhood brainstem malignancy with a 2-year survival rate of <10%. This international survey study aims to evaluate the use of complementary and alternative medicine (CAM) in this patient population. METHODS Parents and physicians of patients with DIPG were asked to participate in a retrospective online survey regarding CAM use during time of illness. RESULTS Between January and May 2020, 120 parents and 75 physicians contributed to the online survey. Most physicians estimated that <50% of their patients used CAM, whereas 69% of the parents reported using CAM to treat their child during time of illness. Cannabis was the most frequently used form of CAM, followed by vitamins and minerals, melatonin, curcumin, and boswellic acid. CAM was mainly used with the intention of direct antitumor effect. Other motivations were to treat side effects of chemotherapy or to increase comfort of the child. Children diagnosed from 2016 onwards were more likely to use CAM (χ2 = 6.08, p = .014). No significant difference was found between CAM users and nonusers based on ethnicity (χ2 = 4.18, p = .382) or country of residence (χ2 = 9.37, p = .154). Almost 50% of the physicians do not frequently ask their patients about possible CAM use. CONCLUSION This survey demonstrates that worldwide, a considerable number of patients with DIPG use CAM. Physicians should be more aware of potential CAM use and actively discuss the topic. In addition, more research is needed to gain knowledge about possible anticancer effects of CAM and (positive/negative) interactions with conventional therapies.
Collapse
Affiliation(s)
- Fatma E El-Khouly
- Pediatric Oncology, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Syed M Adil
- Pediatric Oncology, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Maria Wiese
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Esther Hulleman
- Pediatric Oncology, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - N Harry Hendrikse
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Gertjan J L Kaspers
- Pediatric Oncology, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Christof M Kramm
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Sophie E M Veldhuijzen van Zanten
- Pediatric Oncology, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Dannis G van Vuurden
- Pediatric Oncology, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | |
Collapse
|
43
|
Yu JR, LeRoy G, Bready D, Frenster JD, Saldaña-Meyer R, Jin Y, Descostes N, Stafford JM, Placantonakis DG, Reinberg D. The H3K36me2 writer-reader dependency in H3K27M-DIPG. SCIENCE ADVANCES 2021; 7:eabg7444. [PMID: 34261657 PMCID: PMC8279504 DOI: 10.1126/sciadv.abg7444] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/01/2021] [Indexed: 05/12/2023]
Abstract
Histone H3K27M is a driving mutation in diffuse intrinsic pontine glioma (DIPG), a deadly pediatric brain tumor. H3K27M reshapes the epigenome through a global inhibition of PRC2 catalytic activity and displacement of H3K27me2/3, promoting oncogenesis of DIPG. As a consequence, a histone modification H3K36me2, antagonistic to H3K27me2/3, is aberrantly elevated. Here, we investigate the role of H3K36me2 in H3K27M-DIPG by tackling its upstream catalyzing enzymes (writers) and downstream binding factors (readers). We determine that NSD1 and NSD2 are the key writers for H3K36me2. Loss of NSD1/2 in H3K27M-DIPG impedes cellular proliferation and tumorigenesis by disrupting tumor-promoting transcriptional programs. Further, we demonstrate that LEDGF and HDGF2 are the main readers mediating the protumorigenic effects downstream of NSD1/2-H3K36me2. Treatment with a chemically modified peptide mimicking endogenous H3K36me2 dislodges LEDGF/HDGF2 from chromatin and specifically inhibits the proliferation of H3K27M-DIPG. Our results indicate a functional pathway of NSD1/2-H3K36me2-LEDGF/HDGF2 as an acquired dependency in H3K27M-DIPG.
Collapse
Affiliation(s)
- Jia-Ray Yu
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Gary LeRoy
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Devin Bready
- Department of Neurosurgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Joshua D Frenster
- Department of Neurosurgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Ricardo Saldaña-Meyer
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Ying Jin
- Shared Bioinformatics Core Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Nicolas Descostes
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, New York, NY, USA
- EMBL Rome, Adriano Buzzati-Traverso Campus, Rome, Italy
| | - James M Stafford
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, New York, NY, USA
- Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Dimitris G Placantonakis
- Department of Neurosurgery, New York University Grossman School of Medicine, New York, NY, USA
- Kimmel Center for Stem Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY, USA
- Brain and Spine Tumor Center, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Danny Reinberg
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA.
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
44
|
Erker C, Lane A, Chaney B, Leary S, Minturn JE, Bartels U, Packer RJ, Dorris K, Gottardo NG, Warren KE, Broniscer A, Mark WK, Zhu X, White P, Dexheimer P, Black K, Asher A, DeWire-Shottmiller M, Hansford JR, Gururangan S, Nazarian J, Ziegler DS, Sandler E, Wagner L, Koschmann C, Fuller C, Drissi R, Jones BV, Leach J, Fouladi M. Characteristics of Patients ≥ 10 Years of Age with Diffuse Intrinsic Pontine Glioma: A Report from the International DIPG Registry. Neuro Oncol 2021; 24:141-152. [PMID: 34114629 DOI: 10.1093/neuonc/noab140] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND DIPG generally occurs in young school-age children, although can occur in adolescents and young adults. The purpose of this study was to describe clinical, radiological, pathologic, and molecular characteristics in patients ≥10 years of age with DIPG enrolled in the International DIPG Registry (IDIPGR). METHODS Patients ≥10 years of age at diagnosis enrolled in the IDIPGR with imaging confirmed DIPG diagnosis were included. The primary outcome was overall survival (OS) categorized as long-term survivors (LTS) (≥24 months) or short-term survivors (STS) (<24 months). RESULTS Among 1010 patients, 208 (21%) were ≥10 years of age at diagnosis; 152 were eligible with a median age of 12 years [range 10-26.8]. Median OS was 13 [2-82] months. The 1-, 3- and 5- years OS was 61.9%, 3.7%, and 1.5%, respectively. The 18/152 (11.8%) LTS were more likely to be older (P<0.01) and present with longer symptom duration (P<0.01). Biopsy and/or autopsy were performed in 50 (33%) patients; 77%, 61%, 33%, and 6% of patients tested had H3K27M (H3F3A or HIST1H3B), TP53, ATRX, and ACVR1 mutations/genome alterations, respectively. Two of 18 patients with IDH1 testing were IDH1-mutant and one was a LTS. The presence or absence of H3 alterations did not affect survival. CONCLUSION Patients ≥10 years old with DIPG have a median survival of 13 months. LTS present with longer symptom duration and are likely to be older at presentation compared to STS. ATRX mutation rates were higher in this population than the general DIPG population.
Collapse
Affiliation(s)
- Craig Erker
- Department of Pediatrics, Dalhousie University and IWK Health Center, Halifax, NS, Canada
| | - Adam Lane
- Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center
| | - Brooklyn Chaney
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center
| | - Sarah Leary
- Division of Hematology/Oncology, Seattle Children's Hospital, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Jane E Minturn
- Division of Oncology, Children's Hospital of Philadelphia and Perelman School of Medicine, Philadelphia, PA
| | - Ute Bartels
- Department of Pediatrics, University of Toronto and The Hospital for Sick Children, Toronto, Ontario, Division of Oncology
| | - Roger J Packer
- Department of Neurology, Center for Neuroscience and Behavioral Medicine, Children's National Hospital, Washington, DC
| | - Kathleen Dorris
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Nicholas G Gottardo
- Princess Margaret Hospital for Children and The University of Western Australia, Perth, Western Australia, Australia
| | - Katherine E Warren
- Department of Pediatric Oncology, Dana Farber Cancer Institute/Boston Children's Hospital, Boston, MA, USA
| | - Alberto Broniscer
- Department of Oncology, St. Jude Children's Research Hospital, and Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN
| | - W Kieran Mark
- Department of Pediatrics, Dana-Farber/Boston Children's Cancer and Blood Disorders Center and Harvard Medical School, Boston, MA
| | - Xiaoting Zhu
- Brain Tumor Center, Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.,Department of Electrical Engineering and Computer Science, University of Cincinnati College of Engineering and Applied Science, Cincinnati, OH.,Department of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Peter White
- Department of Biomedical Informatics, Cincinnati Children's Hospital Medical Center and University of Cincinnati
| | - Phillip Dexheimer
- Department of Biomedical Informatics, Cincinnati Children's Hospital Medical Center and University of Cincinnati
| | - Katie Black
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center
| | - Anthony Asher
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center
| | - Mariko DeWire-Shottmiller
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center
| | - Jordan R Hansford
- Royal Children's Hospital, Melbourne; Murdoch Children's Research Institute; Department of Pediatrics, University of Melbourne, Victoria, Australia
| | - Sridharan Gururangan
- Preston A. Wells Center for Brain Tumor Therapy, UF Health Shands Hospital, Gainesville, FL
| | - Javad Nazarian
- Center for Genetic Medicine, Children's National Hospital, Washington D.C., and Department of Oncology, University Children's Hospital, Zurich
| | - David S Ziegler
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW and Kids Cancer Centre, Sydney's Children Hospital, Randwick, Sydney NSW, Australia; and School of Women's and Children's Health, University of New South Wales, Sydney, Australia
| | - Eric Sandler
- Department of Pediatrics, Wolfson Children's Hospital and Nemours Children's Specialty Care, Jacksonville, FL
| | - Lars Wagner
- Division of Pediatric Hematology/Oncology, Kentucky Children's Hospital, University of Kentucky, Lexington, Kentucky
| | - Carl Koschmann
- Department of Pediatrics, C.S. Mott Children's Hospital and University of Michigan School of Medicine, Ann Arbor, MI
| | - Christine Fuller
- Department of Pathology, Upstate Medical University, Syracuse, NY
| | - Rachid Drissi
- Center for Childhood Cancer & Blood Disorders, Nationwide Children's Hospital, Columbus, OH.,The Ohio State University College of Medicine, Columbus, OH
| | - Blaise V Jones
- Division of Radiology, Cincinnati Children's Hospital Medical Center
| | - James Leach
- Division of Radiology, Cincinnati Children's Hospital Medical Center
| | - Maryam Fouladi
- The Ohio State University College of Medicine, Columbus, OH.,Pediatric Neuro-Oncology Program, Nationwide Children's Hospital, Columbus, OH
| |
Collapse
|
45
|
Blattner-Johnson M, Jones DTW, Pfaff E. Precision medicine in pediatric solid cancers. Semin Cancer Biol 2021; 84:214-227. [PMID: 34116162 DOI: 10.1016/j.semcancer.2021.06.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/18/2022]
Abstract
Despite huge advances in the diagnosis and treatment of pediatric cancers over the past several decades, it remains one of the leading causes of death during childhood in developed countries. The development of new targeted treatments for these diseases has been hampered by two major factors. First, the extremely heterogeneous nature of the types of tumors encountered in this age group, and their fundamental differences from common adult carcinomas, has made it hard to truly get a handle on the complexities of the underlying biology driving tumor growth. Second, a reluctance of the pharmaceutical industry to develop products or trials for this population due to the relatively small size of the 'market', and a too-easy mechanism of obtaining waivers for pediatric development of adult oncology drugs based on disease type rather than mechanism of action, led to significant difficulties in getting access to new drugs. Thankfully, the field has now started to change, both scientifically and from a regulatory perspective, in order to address some of these challenges. In this review, we will examine some of the recent insights into molecular features which make pediatric tumors so unique and how these might represent therapeutic targets; highlight ongoing international initiatives for providing comprehensive, personalized genomic profiling of childhood tumors in a clinically-relevant timeframe, and look briefly at where the field of pediatric precision oncology may be heading in future.
Collapse
Affiliation(s)
- Mirjam Blattner-Johnson
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany; Pediatric Glioma Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David T W Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany; Pediatric Glioma Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Elke Pfaff
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany; Pediatric Glioma Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
46
|
Calmon R, Dangouloff-Ros V, Varlet P, Deroulers C, Philippe C, Debily MA, Castel D, Beccaria K, Blauwblomme T, Grevent D, Levy R, Roux CJ, Purcell Y, Saitovitch A, Zilbovicius M, Dufour C, Puget S, Grill J, Boddaert N. Radiogenomics of diffuse intrinsic pontine gliomas (DIPGs): correlation of histological and biological characteristics with multimodal MRI features. Eur Radiol 2021; 31:8913-8924. [PMID: 34003354 DOI: 10.1007/s00330-021-07991-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/10/2021] [Accepted: 04/09/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVES The diffuse intrinsic pontine gliomas (DIPGs) are now defined by the type of histone H3 mutated at lysine 27. We aimed to correlate the multimodal MRI features of DIPGs, H3K27M mutant, with their histological and molecular characteristics. METHODS Twenty-seven treatment-naïve children with histopathologically confirmed DIPG H3K27M mutant were prospectively included. MRI performed prior to biopsy included multi-b-value diffusion-weighted imaging, ASL, and dynamic susceptibility contrast (DSC) perfusion imaging. The ADC and cerebral blood flow (CBF) and blood volume (CBV) were measured at the biopsy site. We assessed quantitative histological data, including microvascular density, nuclear density, and H3K27M-positive nuclear density. Gene expression profiling was also assessed in the samples. We compared imaging and histopathological data according to histone subgroup. We correlated MRI quantitative data with histological data and gene expression. RESULTS H3.1K27M mutated tumors showed higher ADC values (median 3151 μm2/s vs 1741 μm2/s, p = 0.003), and lower perfusion values (DSC-rCBF median 0.71 vs 1.43, p = 0.002, and DSC-rCBV median 1.00 vs 1.71, p = 0.02) than H3.3K27M ones. They had similar microvascular and nuclear density, but lower H3K27M-positive nuclear density (p = 0.007). The DSC-rCBV was positively correlated to the H3K27M-positive nuclear density (rho = 0.74, p = 0.02). ADC values were not correlated with nuclear density nor perfusion values with microvascular density. The expression of gated channel activity-related genes tended to be inversely correlated with ADC values and positively correlated with DSC perfusion. CONCLUSIONS H3.1K27M mutated tumors have higher ADC and lower perfusion values than H3.3K27M ones, without direct correlation with microvascular or nuclear density. This may be due to tissular edema possibly related to gated channel activity-related gene expression. KEY POINTS • H3.1K27M mutant DIPG had higher apparent diffusion coefficient (p = 0.003), lower α (p = 0.048), and lower relative cerebral blood volume (p = 0.02) than H3.3K27M mutant DIPG at their biopsy sites. • Biopsy samples obtained within the tumor's enhancing portion showed higher microvascular density (p = 0.03) than samples obtained outside the tumor's enhancing portion, but similar H3K27M-positive nuclear density (p = 0.84). • Relative cerebral blood volume measured at the biopsy site was significantly correlated with H3K27M-positive nuclear density (rho = 0.74, p = 0.02).
Collapse
Affiliation(s)
- Raphaël Calmon
- Pediatric Radiology Department, AP-HP, Hôpital Universitaire Necker-Enfants Malades, 149 rue de Sèvres, F-75015, Paris, France.,Université de Paris, INSERM ERL UA10, F-75015, Paris, France.,Université de Paris, UMR 1163, Institut Imagine, F-75015, Paris, France
| | - Volodia Dangouloff-Ros
- Pediatric Radiology Department, AP-HP, Hôpital Universitaire Necker-Enfants Malades, 149 rue de Sèvres, F-75015, Paris, France. .,Université de Paris, INSERM ERL UA10, F-75015, Paris, France. .,Université de Paris, UMR 1163, Institut Imagine, F-75015, Paris, France.
| | - Pascale Varlet
- Neuropathology Department, Sainte-Anne Hospital, F-75014, Paris, France.,Université de Paris, INSERM U894, IMA BRAIN, F-75014, Paris, France
| | | | - Cathy Philippe
- Université Paris-Saclay, Neurospin, Institut Joliot, CEA, Gif-sur-Yvette, France
| | | | - David Castel
- Université Paris-Saclay, UMR8203, CNRS, F-94805, Villejuif, France
| | - Kevin Beccaria
- Pediatric Neurosurgery Department, AP-HP, Hôpital Universitaire Necker-Enfants Malades, F-75015, Paris, France.,Université de Paris, F-75015, Paris, France
| | - Thomas Blauwblomme
- Pediatric Neurosurgery Department, AP-HP, Hôpital Universitaire Necker-Enfants Malades, F-75015, Paris, France.,Université de Paris, F-75015, Paris, France
| | - David Grevent
- Pediatric Radiology Department, AP-HP, Hôpital Universitaire Necker-Enfants Malades, 149 rue de Sèvres, F-75015, Paris, France.,Université de Paris, INSERM ERL UA10, F-75015, Paris, France.,Université de Paris, UMR 1163, Institut Imagine, F-75015, Paris, France
| | - Raphael Levy
- Pediatric Radiology Department, AP-HP, Hôpital Universitaire Necker-Enfants Malades, 149 rue de Sèvres, F-75015, Paris, France.,Université de Paris, INSERM ERL UA10, F-75015, Paris, France.,Université de Paris, UMR 1163, Institut Imagine, F-75015, Paris, France
| | - Charles-Joris Roux
- Pediatric Radiology Department, AP-HP, Hôpital Universitaire Necker-Enfants Malades, 149 rue de Sèvres, F-75015, Paris, France.,Université de Paris, INSERM ERL UA10, F-75015, Paris, France.,Université de Paris, UMR 1163, Institut Imagine, F-75015, Paris, France
| | - Yvonne Purcell
- Radiology Department, Fondation Rothschild, F-75019, Paris, France
| | - Ana Saitovitch
- Université de Paris, INSERM ERL UA10, F-75015, Paris, France.,Université de Paris, UMR 1163, Institut Imagine, F-75015, Paris, France
| | - Monica Zilbovicius
- Université de Paris, INSERM ERL UA10, F-75015, Paris, France.,Université de Paris, UMR 1163, Institut Imagine, F-75015, Paris, France
| | - Christelle Dufour
- Université Paris-Saclay, UMR8203, CNRS, F-94805, Villejuif, France.,Department of Pediatric and Adolescent Oncology, Institut Gustave Roussy, F-94805, Villejuif, France
| | - Stéphanie Puget
- Pediatric Neurosurgery Department, AP-HP, Hôpital Universitaire Necker-Enfants Malades, F-75015, Paris, France.,Université de Paris, F-75015, Paris, France
| | - Jacques Grill
- Université Paris-Saclay, UMR8203, CNRS, F-94805, Villejuif, France.,Department of Pediatric and Adolescent Oncology, Institut Gustave Roussy, F-94805, Villejuif, France
| | - Nathalie Boddaert
- Pediatric Radiology Department, AP-HP, Hôpital Universitaire Necker-Enfants Malades, 149 rue de Sèvres, F-75015, Paris, France.,Université de Paris, INSERM ERL UA10, F-75015, Paris, France.,Université de Paris, UMR 1163, Institut Imagine, F-75015, Paris, France
| |
Collapse
|
47
|
A phase I/II study of bevacizumab, irinotecan and erlotinib in children with progressive diffuse intrinsic pontine glioma. J Neurooncol 2021; 153:263-271. [PMID: 33963476 PMCID: PMC8211596 DOI: 10.1007/s11060-021-03763-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/20/2021] [Indexed: 12/26/2022]
Abstract
INTRODUCTION This study investigates the safety, tolerability, and preliminary efficacy of combined treatment with VEGF inhibitor bevacizumab, topoisomerase I inhibitor irinotecan, and EGFR inhibitor erlotinib in children with progressive diffuse intrinsic pontine glioma (DIPG). METHODS Biweekly bevacizumab (10 mg/kg) and irinotecan (125 mg/m2) were combined with daily erlotinib. Two cohorts received increasing doses of erlotinib (65 and 85 mg/m2) following a 3 + 3 dose-escalation schedule, until disease progression with a maximum of one year. Dose-limiting toxicities (DLT) were monitored biweekly. Secondary progression free survival (sPFS) and overall survival (OS) were determined based on clinical and radiological response measurements. Quality of life (QoL) during treatment was also assessed. RESULTS Between November 2011 and March 2018, nine patients with disease progression after initial radiotherapy were enrolled. Median PFS at start of the study was 7.3 months (range 3.5-10.0). In the first dose cohort, one patient experienced a DLT (grade III acute diarrhea), resulting in enrollment of three additional patients in this cohort. No additional DLTs were observed in consecutive patients receiving up to a maximum dose of 85 mg/m2. Median sPFS was 3.2 months (range 1.0-10.9), and median OS was 13.8 months (range 9.3-33.0). Overall QoL was stable during treatment. CONCLUSIONS Daily erlotinib is safe and well tolerated in doses up to 85 mg/m2 when combined with biweekly bevacizumab and irinotecan in children with progressive DIPG. Median OS of the study patients was longer than known form literature.
Collapse
|
48
|
Abstract
Pediatric gliomas are biologically distinct from adult gliomas. Although recent literature uncovered new genetic alterations, the prognostic implications of these discoveries are still unclear. This article provides an update on the histologic and molecular features with prognostic and/or therapeutic implications in pediatric gliomas.
Collapse
Affiliation(s)
- Jared Ahrendsen
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA 02115, USA
| | - Sanda Alexandrescu
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Bader 104, Boston, MA 02115, USA.
| |
Collapse
|
49
|
Leach JL, Roebker J, Schafer A, Baugh J, Chaney B, Fuller C, Fouladi M, Lane A, Doughman R, Drissi R, DeWire-Schottmiller M, Ziegler DS, Minturn JE, Hansford JR, Wang SS, Monje-Deisseroth M, Fisher PG, Gottardo NG, Dholaria H, Packer R, Warren K, Leary SES, Goldman S, Bartels U, Hawkins C, Jones BV. MR imaging features of diffuse intrinsic pontine glioma and relationship to overall survival: report from the International DIPG Registry. Neuro Oncol 2021; 22:1647-1657. [PMID: 32506137 DOI: 10.1093/neuonc/noaa140] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND This study describes imaging features of diffuse intrinsic pontine glioma (DIPG) and correlates with overall survival (OS) and histone mutation status in the International DIPG Registry (IDIPGR). METHODS Four hundred cases submitted to the IDIPGR with a local diagnosis of DIPG and baseline MRI were evaluated by consensus review of 2 neuroradiologists; 43 cases were excluded (inadequate imaging or alternative diagnoses). Agreement between reviewers, association with histone status, and univariable and multivariable analyses relative to OS were assessed. RESULTS On univariable analysis imaging features significantly associated with worse OS included: extrapontine extension, larger size, enhancement, necrosis, diffusion restriction, and distant disease. On central review, 9.5% of patients were considered not to have DIPG. There was moderate mean agreement of MRI features between reviewers. On multivariable analysis, chemotherapy, age, and distant disease were predictors of OS. There was no difference in OS between wild-type and H3 mutated cases. The only imaging feature associated with histone status was the presence of ill-defined signal infiltrating pontine fibers. CONCLUSIONS Baseline imaging features are assessed in the IDIPGR. There was a 9.5% discordance in DIPG diagnosis between local and central review, demonstrating need for central imaging confirmation for prospective trials. Although several imaging features were significantly associated with OS (univariable), only age and distant disease were significant on multivariable analyses. There was limited association of imaging features with histone mutation status, although numbers are small and evaluation exploratory.
Collapse
Affiliation(s)
- James L Leach
- Department of Radiology and Medical Imaging, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - James Roebker
- Department of Radiology and Medical Imaging, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Austin Schafer
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Joshua Baugh
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Neuro-oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Brooklyn Chaney
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Christine Fuller
- Department of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Maryam Fouladi
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Adam Lane
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Renee Doughman
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Rachid Drissi
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | | | - Jane E Minturn
- Division of Oncology, Children's Hospital of Philadelphia, Pennsylvania
| | - Jordan R Hansford
- Children's Cancer Centre, Royal Children's Hospital; Murdoch Children's Research Institute; University of Melbourne, Melbourne, Australia
| | - Stacie S Wang
- Children's Cancer Centre, Royal Children's Hospital; Murdoch Children's Research Institute; University of Melbourne, Melbourne, Australia
| | | | - Paul G Fisher
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, California
| | | | - Hetal Dholaria
- Department of Oncology, Perth Children's Hospital, Perth, AU
| | - Roger Packer
- Division of Oncology, Children's National Medical Center, Washington, DC
| | - Katherine Warren
- Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Harvard Cancer Center, Boston Massachusetts
| | - Sarah E S Leary
- Cancer and Blood Disorders Center, Seattle Children's, Seattle, Washington
| | - Stewart Goldman
- Division of Oncology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Ute Bartels
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, CA
| | - Cynthia Hawkins
- Division of Pathology, The Hospital for Sick Children, Toronto, CA
| | - Blaise V Jones
- Department of Radiology and Medical Imaging, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
50
|
Cacciotti C, Liu KX, Haas-Kogan DA, Warren KE. Reirradiation practices for children with diffuse intrinsic pontine glioma. Neurooncol Pract 2021; 8:68-74. [PMID: 33664971 DOI: 10.1093/nop/npaa063] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Background Diffuse intrinsic pontine gliomas (DIPGs) are a leading cause of brain tumor deaths in children. Current standard of care includes focal radiation therapy (RT). Despite clinical improvement in most patients, the effect is temporary and median survival is less than 1 year. The use and benefit of reirradiation have been reported in progressive DIPG, yet standardized approaches are lacking. We conducted a survey to assess reirradiation practices for DIPG in North America. Methods A 14-question REDCap survey was disseminated to 396 North American physicians who care for children with CNS tumors. Results The response rate was 35%. Participants included radiation-oncologists (63%; 85/135) and pediatric oncologists/neuro-oncologists (37%; 50/135). Most physicians (62%) treated 1 to 5 DIPG patients per year, with 10% treating more than 10 patients per year. Reirradiation was considered a treatment option by 88% of respondents. Progressive disease and worsening clinical status were the most common reasons to consider reirradiation. The majority (84%) surveyed considered reirradiation a minimum of 6 months following initial RT. Doses varied, with median total dose of 2400 cGy (range, 1200-6000 cGy) and fraction size of 200 cGy (range, 100-900 cGy). Concurrent use of systemic agents with reirradiation was considered in 46%, including targeted agents (37%), biologics (36%), or immunotherapy (25%). One-time reirradiation was the most common practice (71%). Conclusion Although the vast majority of physicians consider reirradiation as a treatment for DIPG, total doses and fractionation varied. Further clinical trials are needed to determine the optimal radiation dose and fractionation for reirradiation in children with progressive DIPG.
Collapse
Affiliation(s)
- Chantel Cacciotti
- Dana Farber/Boston Children's Cancer and Blood Disorder Center, Boston, Massachusetts
| | - Kevin X Liu
- Department of Radiation-Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Daphne A Haas-Kogan
- Department of Radiation-Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Katherine E Warren
- Dana Farber/Boston Children's Cancer and Blood Disorder Center, Boston, Massachusetts
| |
Collapse
|