1
|
van Ommen F, van Genechten T, Willemsen-Bosman ME, Peters M, Seravalli E, van der Lugt J, Nievelstein RAJ, Mueller S, Hulleman E, van Vuurden DG, Kranendonk MEG, Hoving EW, Hoeben BAW, Janssens GO. Gross tumor volume increase and need for adaptive radiotherapy in pediatric-type diffuse high-grade glioma of the midline structures. Radiother Oncol 2025; 207:110873. [PMID: 40174703 DOI: 10.1016/j.radonc.2025.110873] [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: 12/09/2024] [Revised: 03/18/2025] [Accepted: 03/25/2025] [Indexed: 04/04/2025]
Abstract
INTRODUCTION Current pediatric-type diffuse high-grade glioma radiotherapy protocols apply a 1.0 cm clinical target volume (CTV) margin around the gross tumor volume (GTV). However, in adults with glioblastoma, large variations in GTV are observed during radiotherapy. The study aimed to map the GTV variation during a 6-week course of radiotherapy using repeated MR-imaging and to evaluate the need for plan adaptation. Also, the relation between GTV increase and time to disease progression (TTP) was assessed. MATERIAL AND METHODS Patients with newly diagnosed diffuse midline glioma or diffuse pediatric-type high-grade glioma of the midline structures undergoing a 6-week radiotherapy course, were eligible for inclusion. MRI scans were performed in the pre-treatment phase (MRI0), and at fraction 10 + 20 (rMRI10/rMRI20). On all scans, GTV was delineated. An increase was defined as a >5 % increase of GTV between scans. The need for treatment plan adaptation was based on dosimetric and visual criteria. GTV increase was compared to TTP. RESULTS Twenty patients were eligible. In 12/20 patients, a GTV increase was observed at rMR10/rMR20, more specifically in 6/11 pontine and 6/9 non-pontine tumors. Combining dosimetric criteria and visual inspection, 20 plan adaptations in 14 patients were required. The TTP (range: 1.6-17.6 months) was not significantly different between the group with (median 8.1 months) versus without a GTV increase (median 7.6 months; p = 0.66). CONCLUSION Repeated imaging demonstrated a GTV increase in 60 % of patients and plan adaptation in 70 %. When applying CTV margins of 1.0 cm, plan adaptation is recommended to ensure adequate radiotherapy treatment.
Collapse
Affiliation(s)
- Fasco van Ommen
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Toon van Genechten
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pediatric Oncology, University Hospital Antwerp, Antwerp, Belgium
| | | | - Max Peters
- Radiotherapiegroep Deventer, Deventer, the Netherlands
| | - Enrica Seravalli
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Rutger A J Nievelstein
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Radiology & Nuclear Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Sabine Mueller
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Neurology, Neurosurgery and Pediatrics, University of California San Francisco, San Francisco, United States
| | - Esther Hulleman
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | | | - Eelco W Hoving
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Bianca A W Hoeben
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, the Netherlands; Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Geert O Janssens
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, the Netherlands; Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| |
Collapse
|
2
|
Rathi A, Choudhari AK, Chatterjee A, Sahay A, Bhattacharya K, Sahu A, Puranik AD, Shah A, Dasgupta A, Gupta T, Epari S. Spinal Diffuse Midline Glioma H3 K27M-Altered: Report of a Rare Tumor with Extracranial Skeletal Metastases and Review of Literature. Int J Surg Pathol 2025; 33:907-915. [PMID: 39376094 DOI: 10.1177/10668969241286243] [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] [Indexed: 10/09/2024]
Abstract
Diffuse midline glioma, H3 K27-altered is a rare and aggressive pediatric brain tumor with a grim prognosis. Diffuse midline glioma is characterized by specific molecular alterations, including H3 K27 mutations, and involves deep midline structures such as the brainstem, cerebellum, spinal cord, and thalamus. These tumors present with a classic triad of symptoms and have limited surgical options due to their challenging locations. Extra-neural metastases are an unusual occurrence in diffuse midline glioma and have been rarely described. Here we report a 17-year-old girl with spinal diffuse midline glioma, H3 K27M-mutant, who presented with multiple metastatic osseous lesions confirmed on biopsy of the thoracic vertebral lesion. Due to the rapid disease progression, the patient was recommended palliative therapy. Extra-neural metastases in diffuse midline glioma are rare, with only 16 reported patients, and no standard therapy exists. An accurate and early diagnosis is necessary to develop a personalized plan of treatment. Further research is needed to gain insights into the molecular pathology of diffuse midline glioma, H3 K27-altered, and improve the quality of life and the outcome of patients with this deadly disease.
Collapse
Affiliation(s)
- Aditi Rathi
- Department of Pathology, Tata Memorial Hospital and ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Amit Kumar Choudhari
- Department of Radiology, Tata Memorial Hospital and ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Abhishek Chatterjee
- Department of Nuclear Medicine, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Ayushi Sahay
- Department of Pathology, Tata Memorial Hospital and ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Kajari Bhattacharya
- Department of Radiology, Tata Memorial Hospital and ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Arpita Sahu
- Department of Radiology, Tata Memorial Hospital and ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Ameya D Puranik
- Department of Nuclear Medicine, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Aekta Shah
- Department of Pathology, Tata Memorial Hospital and ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Archya Dasgupta
- Department of Radiation Oncology, Tata Memorial Hospital and ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Tejpal Gupta
- Department of Radiation Oncology, Tata Memorial Hospital and ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Sridhar Epari
- Department of Pathology, Tata Memorial Hospital and ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
3
|
Deng Y, Liu J, Pu Z, Wang Y, Li T, Jiang Z, Xie L, Zhang X, Chen Y, Yang M, Du C, Hao S, Ji N, Zhuang Z, Feng J, Zhang L. Targeting the HLA-E-NKG2A axis in combination with MS-275 enhances NK cell-based immunotherapy against DMG. J Exp Clin Cancer Res 2025; 44:133. [PMID: 40296045 PMCID: PMC12039099 DOI: 10.1186/s13046-025-03390-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Diffuse midline glioma (DMG) is an aggressive pediatric brain tumor with limited treatment options. Although natural killer (NK) cell-based immunotherapy is promising, its efficacy remains limited, necessitating strategies to enhance NK cell cytotoxicity. Histone deacetylase (HDAC) inhibition demonstrate potential to enhance NK-mediated killing. However, the combination of HDAC inhibitors and NK cell therapy for DMG remains unexplored. METHODS Patient-derived DMG cell lines and orthotopic mouse models were used to evaluate the effects of the class I HDAC inhibitor MS-275 on cytotoxicity. NK cell-mediated lysis was measured using both luciferase and calcein AM-based assays. The downstream signaling pathways affected by MS-275 were investigated via RNA-seq, CUT&Tag assay, RT‒qPCR, and chromatin immunoprecipitation with qPCR. RESULTS Based on bioinformatic analysis, class I HDACs are identified as therapeutic targets in DMG. The corresponding HDAC inhibitor, MS-275 upregulated NK cell-mediated cytotoxicity pathway through GSEA analysis. Pretreating DMG cells with MS-275 elevated NK cell ligand gene expression and enhanced NK cell-induced lysis. In addition to NK-activating ligands, MS-275 elevated the NK-inhibitory ligand HLA-E, thereby enhancing the efficacy of immunotherapies targeting the NKG2A-HLA-E axis. Mechanistically, MS-275 increased HLA-E expression by promoting STAT3 acetylation at lysine 685. Combining MS-275 with NK cell therapy and blockade of the NKG2A-HLA-E axis extended overall survival in orthotopic mouse models. CONCLUSIONS This study is the first to demonstrate that HDAC inhibition enhances NK cell-mediated cytotoxicity in DMG. Combining HDAC inhibition with NK cell therapy represents a promising therapeutic strategy for treating DMG by targeting NKG2A-HLA-E axis.
Collapse
Affiliation(s)
- Yuxuan Deng
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinqiu Liu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zhuonan Pu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yi Wang
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tian Li
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhuang Jiang
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Luyang Xie
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaoli Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - YingDan Chen
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Mingxu Yang
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chao Du
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuyu Hao
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Nan Ji
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhengping Zhuang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jie Feng
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| | - Liwei Zhang
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| |
Collapse
|
4
|
Luo XH, Zhu Y, Duan XQ, Peng W, Pei CX, Yang L, Li Q, Zhao M, Wang L. Histone HIST1 genes and tumor-infiltrating lymphocytes in a child with γδ T cell acute lymphoblastic leukemia by single-cell sequencing. J Leukoc Biol 2025; 117:qiaf022. [PMID: 39973604 DOI: 10.1093/jleuko/qiaf022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 12/31/2024] [Accepted: 02/18/2025] [Indexed: 02/21/2025] Open
Abstract
γδ T cell acute lymphoblastic leukemia (γδ T-ALL) represents a rare subset of T-ALL and is correlated with high rates of induction failure, relapse, and increased mortality. γδ T-ALL lacks a biologically informed framework for guiding its classification and treatment strategies. In this report, we detail a case of child with γδ T-ALL who underwent induction chemotherapy and intensification treatment, followed by haploidentical hematopoietic stem cell transplantation. The patient achieved a clinical complete remission and remains minimal residual disease negative with chidamide maintenance post-transplantation. Single-cell RNA sequencing revealed a connection between histone HIST1 genes and γδ T-ALL and identified potential effector functions of γδ T cells in combating this leukemia. This case carries significant implications for managing γδ T-ALL, highlighting the relationship between histone modification patterns and γδ tumor-infiltrating lymphocytes in γδ T-ALL cells for developing novel therapeutic approaches.
Collapse
Affiliation(s)
- Xiao-Hua Luo
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Yan Zhu
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Xiao-Qin Duan
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Wen Peng
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Cai-Xia Pei
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Li Yang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Qing Li
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Min Zhao
- Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing 400016, China
| | - Lan Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing 400016, China
| |
Collapse
|
5
|
Jia H, Wang K, Zhang P, Zhang M, Mai Y, Chu C, Yin X, Fan L, Zhang L. Cortical thickness deviations as biomarker for subtyping and prognosis in pediatric brainstem tumors. Sci Rep 2025; 15:13132. [PMID: 40240399 PMCID: PMC12003633 DOI: 10.1038/s41598-025-95017-7] [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: 09/24/2024] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Brainstem tumors exert profound effects on cortical organization and functionality across the whole brain. However, the precise implications of changes in cortical thickness (CTh) for patient stratification and prognostic assessment remain unclear. Our study seeks to address these gaps and provide clearer insights into the distant impact of brainstem tumors. This study involved 124 pediatric patients with brainstem tumors and 849 healthy controls. Using CAT12 segmentation on 3D T1-weighted MRI scans and Gaussian process regression modeling, we established a normative CTh model from healthy data. CTh deviations of patients were quantified and clustered, revealing two distinct subtypes: Subtype 1 with extremely positive deviations and Subtype 2 with extremely negative deviations, correlating with better survival. Kaplan-Meier analysis confirmed significant survival differences between these subtypes. Additionally, a greater number of brain regions with positive CTh deviations was found to correlate with larger tumor volumes. These findings suggest that CTh deviation is a non-invasive imaging marker, facilitating patient subtyping and survival prediction. These insights equip clinicians to tailor treatment plans and establishes a valuable precision medical tool for clinical evaluation and monitoring.
Collapse
Affiliation(s)
- Heyuan Jia
- School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, China
- Institute of Large-scale Scientific Facility and Centre for Zero Magnetic Field Science, Beihang University, Beijing, China
| | - Kaikai Wang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Peng Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Mingxin Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yiying Mai
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Congying Chu
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xuntao Yin
- Department of Radiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Lingzhong Fan
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, 266000, China.
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
| |
Collapse
|
6
|
Wang K, Graziano G, Ceisel A, Xiao H, Banerjee S, Yu Y, Galanternik MV, Weinstein BM, Eberhart CG, Mumm J, Raabe E. Developing a zebrafish xenograft model of diffuse midline glioma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646163. [PMID: 40236039 PMCID: PMC11996398 DOI: 10.1101/2025.03.31.646163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Diffuse midline glioma (DMG) is a highly aggressive brain tumor that predominantly affects children. Conventional treatments such as radiation therapy can control progression for a time, but DMG kills nearly 100 percent of patients. Although murine models have provided critical insights into the biology of DMG and in assessing new therapeutic strategies, they are not suitable for high-throughput screening to identify and profile novel therapies due to technical challenges, ethical considerations and high cost. Zebrafish ( Danio rerio ) is an established vertebrate model for large-scale drug screening, and zebrafish have demonstrated the ability to replicate the key biological and pathlogical aspects of human malignancies. Here, we developed a novel method for transplanting human DMG cells into large numbers of zebrafish embyros to speed the assessment of anti-tumor drug efficacy in vivo and thereby facilitate the development of novel therapeutics for clinical translation. We transplanted red fluorescent protein (RFP)-labeled, patient-derived DMG cell lines into zebrafish blastulas. Remarkably, many DMG cells migrate into the developing brain and are present in the midline of the brain 24 hours after blastula injection. Tumor cell burden was monitored by measuring RFP fluorescence intensity changes over time. Time-course images of transplanted tumor cell volumes were acquired, and the interactions between transplanted DMG cells and microglial cells were further analyzed using Imaris software. We have developed a simple and rapid transplantation protocol to establish a zebrafish xenograft model of DMG. Our method involves transplanting DMG cells into the blastula stage (1000 cell stage) of zebrafish embryos, which does not require complex surgical techniques. This approach allows for the transplantation of hundreds of embryos per hour, significantly increasing the efficiency of creating DMG zebrafish xenografts that are suitable for high-throughput drug and gene discovery screens.
Collapse
|
7
|
Zahid S, Bashir F, Mustansir A, Minhas K, Qureshi BM, Hilal K, Enam SA, Bouffet E, Mushtaq N. Frequency and outcomes of midline gliomas in a tertiary care hospital in Pakistan: a retrospective study. Childs Nerv Syst 2025; 41:148. [PMID: 40169441 DOI: 10.1007/s00381-025-06811-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/24/2025] [Indexed: 04/03/2025]
Abstract
INTRODUCTION Midline structures in the central nervous system include the thalamus, brainstem, and spinal cord. Within the midline tumors, Diffuse midline gliomas (DMGs) and diffuse intrinsic pontine gliomas (DIPG) have a poor prognosis. DMGs are inclusive of all diffuse intrinsic pontine gliomas (DIPG), previously usually used for only pontine gliomas, to emphasize that these lesions are not solely centered in the pons/brainstem. In this retrospective review, we aim to report the frequency and outcomes of midline gliomas amongst all midline tumors in a tertiary care setup. METHODS Data were collected retrospectively from the medical records at Aga Khan University Hospital between 2013 and 2023. All patients aged 18 and younger with tumors in midline locations were reviewed, and 102 patients were included. A few tumor samples were also sent to SickKids, Toronto, for molecular testing. RESULTS Our cohort represents 102 patients with midline tumors, a median age of 11 years (interquartile range (IQR): 7.75-15 years), and a similar male-to-female ratio. Most patients presented with limb weakness and headache (median duration: 1.5 months, IQR: 1-4 months). The most common site of tumors was the brainstem, followed by the spine and thalamus. Sixty-six patients had surgery: 2 DIPGs, 15 low-grade gliomas, 13 ependymomas, 8 high-grade gliomas, 12 diffuse midline glioma, and 16 other tumors were identified. All of the patients diagnosed with DMG had H3K27 alteration on immunohistochemistry. Thirty-six patients were diagnosed via radiology: 33 DIPG and 3 tectal plate glioma. Only 10 patients received chemotherapy, and radiation therapy was given to 24 patients. Overall survival for all midline tumors was 53.9%, with 47 events. CONCLUSION Our study depicts poor survival outcomes at one year of patients diagnosed with DMG (16.7%) and DIPG (14.3%) amongst all midline tumors, regardless of radiation therapy or concurrent chemoradiotherapy. To improve the care and survival of all midline tumors, there is a dire need for affordable diagnostic techniques in specialized centers across low- and middle-income countries.
Collapse
Affiliation(s)
- Soha Zahid
- Section of Pediatric-Oncology, Department of Oncology, Aga Khan University Hospital, Stadium Road, P.O. Box 3500, Karachi, 74800, Pakistan.
| | - Farrah Bashir
- Section of Pediatric-Oncology, Department of Oncology, Aga Khan University Hospital, Stadium Road, P.O. Box 3500, Karachi, 74800, Pakistan
| | - Ali Mustansir
- Medical College, Aga Khan University Hospital, Karachi, Pakistan
| | - Khurram Minhas
- Department of Pathology and Laboratory Medicine, Aga Khan University Hospital Karachi, Karachi, Pakistan
| | - Bilal Mazhar Qureshi
- Section of Pediatric-Oncology, Department of Oncology, Aga Khan University Hospital, Stadium Road, P.O. Box 3500, Karachi, 74800, Pakistan
| | - Kiran Hilal
- Department of Radiology, Aga Khan University Hospital, Karachi, Pakistan
| | - Syed Ather Enam
- Department of Surgery, Aga Khan University Hospital, Karachi, Pakistan
| | - Eric Bouffet
- Section of Pediatric-Oncology, Department of Oncology, Aga Khan University Hospital, Stadium Road, P.O. Box 3500, Karachi, 74800, Pakistan
- The Hospital for Sick Children, Toronto, Canada
| | - Naureen Mushtaq
- Section of Pediatric-Oncology, Department of Oncology, Aga Khan University Hospital, Stadium Road, P.O. Box 3500, Karachi, 74800, Pakistan
| |
Collapse
|
8
|
Marastoni E, Ammendola S, Rossi S, Giovannoni I, Broggi G, Masotto B, Feletti A, Barresi V. H3 K27M mutation in rosette-forming glioneuronal tumors: a potential diagnostic pitfall. Virchows Arch 2025; 486:781-789. [PMID: 38233563 PMCID: PMC12018613 DOI: 10.1007/s00428-024-03739-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/18/2023] [Accepted: 01/11/2024] [Indexed: 01/19/2024]
Abstract
According to the fifth edition of the World Health Organization (WHO) classification of tumors of the central nervous system (CNS), diffuse midline glioma H3 K27-altered is a grade 4 infiltrative glioma that arises from midline anatomical structures and is characterized by the loss of H3 K27me3 and co-occurring H3 K27M mutation or EZHIP overexpression. However, the H3 K27M mutation has also been observed in circumscribed gliomas and glioneuronal tumors arising in midline anatomical structures, which may result in diagnostic pitfalls.Rosette-forming glioneuronal tumor (RGNT) is a CNS WHO grade 1 neoplasm that histologically features neurocytic and glial components and originates in midline anatomical structures.This study aimed to assess whether RGNTs, similar to other midline tumors, may exhibit immunohistochemical loss of H3 K27me3 and harbor the H3 K27M mutation.All seven analyzed RGNTs displayed immunohistochemical loss of H3 K27me3 in all tumor cells or H3 K27me3 mosaic immunostaining. In one case, H3 K27me3 loss was associated with the H3 K27M mutation, whereas the other six cases did not exhibit any H3 mutations or EZHIP overexpression. During a follow-up period of 23 months, the H3 K27M-mutant case remained unchanged in size despite partial resection, indicating that the H3 mutation may not confer higher biological aggressiveness to RGNT.The immunohistochemical loss of H3 K27me3 co-occurring with the H3 K27M mutation may result in the potential misdiagnosis of RGNT, especially in cases of small biopsy specimens consisting of only the glial component.
Collapse
Affiliation(s)
- Elena Marastoni
- Department of Diagnostics and Public Health, University of Verona, Policlinico G.B. Rossi, P.le L.A. Scuro, 10, 37138, Verona, Italy
| | - Serena Ammendola
- Department of Diagnostics and Public Health, University of Verona, Policlinico G.B. Rossi, P.le L.A. Scuro, 10, 37138, Verona, Italy
| | - Sabrina Rossi
- Unit of Anatomic Pathology, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | | | - Giuseppe Broggi
- Department of Medical and Surgical Sciences and Advanced Technologies, G.F. Ingrassia, Anatomic Pathology, University of Catania, Catania, Italy
| | - Barbara Masotto
- Unit of Cranial Posterior Fossa Surgery, University and Hospital Trust of Verona, Verona, Italy
| | - Alberto Feletti
- Department of Neurosciences, University of Verona, Verona, Italy
| | - Valeria Barresi
- Department of Diagnostics and Public Health, University of Verona, Policlinico G.B. Rossi, P.le L.A. Scuro, 10, 37138, Verona, Italy.
| |
Collapse
|
9
|
Saito R, Kanamori M, Arakawa Y, Mineharu Y, Aihara Y, Chiba K, Kumabe T, Shibahara I, Sonoda Y, Matsuda K, Kinoshita M, Sato A, Takahashi F, Tominaga T. A Multicenter Phase II Trial of Nimustine Hydrochloride Administered via Convection-Enhanced Delivery in Children With DIPG. Cancer Sci 2025. [PMID: 40150841 DOI: 10.1111/cas.70054] [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: 12/31/2024] [Revised: 02/28/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a very challenging-to-treat pediatric malignant tumor, with a median survival time of < 12 months. Convection-enhanced delivery (CED) allows for direct drug administration into the tumor site, showing potential as a novel therapeutic approach. This study evaluated the efficacy of CED of nimustine hydrochloride (ACNU) in children with DIPG. This phase 2, single-arm, multicenter study enrolled patients aged 3-21 years and diagnosed with DIPG. The investigational treatment commenced 1 month after completing radiotherapy (local 50-60 Gy). The treatment involved stereotactic brain surgery for catheter placement, followed by ACNU administration via a CED catheter at a concentration of 0.75 mg/mL for 2-3 days until a cumulative dose of 7 (±0.3) mL was achieved. The primary endpoint was the 1-year survival rate. From April 2018 to March 2020, 21 children were enrolled in the trial and treated, with 20 evaluable for the primary endpoint. The 1-year survival rate from the start of radiotherapy was 60%, and the median survival time was 15 months. The response rate was analyzed in 20 patients, with one complete response (CR), six partial responses (PR), nine stable diseases, and four progressive diseases, resulting in a response rate of 35% (CR + PR). The CED of ACNU in the brainstem of children with DIPG after radiotherapy appears to be an effective therapeutic strategy. This approach warrants further development as a treatment for children with DIPG. This study is registered with jRCT (No. jRCT2021190003).
Collapse
Affiliation(s)
- Ryuta Saito
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Masayuki Kanamori
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Neurosurgery, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Yoshiki Arakawa
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
- Department of Neurosurgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Yohei Mineharu
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Yasuo Aihara
- Department of Neurosurgery, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Kentaro Chiba
- Department of Neurosurgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Toshihiro Kumabe
- Department of Neurosurgery, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Ichiyo Shibahara
- Department of Neurosurgery, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Yukihiko Sonoda
- Department of Neurosurgery, Yamagata University, Yamagata, Japan
| | | | - Manabu Kinoshita
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Aya Sato
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Fumiaki Takahashi
- Department of Information Science, Iwate Medical University, Iwate, Japan
| | - Teiji Tominaga
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
10
|
Kahvecioglu A, Cengiz M, Burca Aydin G, Tezer Kutluk M, Coban Cifci G, Yazici G. Re-irradiation in progressive diffuse infiltrative pontine glioma in children and young adults. Strahlenther Onkol 2025:10.1007/s00066-025-02394-z. [PMID: 40131412 DOI: 10.1007/s00066-025-02394-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 03/05/2025] [Indexed: 03/27/2025]
Abstract
PURPOSE This study aims to assess oncological outcomes in children and young adults with diffuse infiltrative pontine glioma (DIPG) who have progressed after initial radiotherapy (RT), with an emphasis on the role of re-irradiation. METHODS Data from 33 patients aged 25 years or younger with progressive disease after initial RT were retrospectively analyzed. RESULTS The median age at diagnosis was 8 years (range 4-24 years), and the median initial RT dose was 54 Gy (range 39-54 Gy). The median time between initial RT and progression was 8 months (range 3-40 months). In addition to systemic therapy, 15 patients (46%) received re-irradiation due to progression, with a median dose of 23.4 Gy (range 19.8-36 Gy), while 18 patients (54%) were treated with systemic therapy alone. In patients who received re-irradiation after progression, the 1‑year post-progression overall survival (OS) was significantly higher compared to those treated with systemic therapy alone (27% vs. 0%, p = 0.01). Among the 15 re-irradiated patients, 9 out of 12 with available data (75%) showed improvement in neurological symptoms following re-irradiation. No patient exhibited acute or late RT-related ≥ grade 3 toxicity. CONCLUSION Palliative re-irradiation in children and young adults with progressive DIPG after initial RT provides an approximately 3‑month OS benefit and clinical improvement without significant toxicity and should be considered as a standard-of-care approach.
Collapse
Affiliation(s)
- Alper Kahvecioglu
- Faculty of Medicine, Department of Radiation Oncology, Hacettepe University, Sihhiye, 06100, Ankara, Turkey
| | - Mustafa Cengiz
- Faculty of Medicine, Department of Radiation Oncology, Hacettepe University, Sihhiye, 06100, Ankara, Turkey
| | - Guzide Burca Aydin
- Faculty of Medicine, Department of Pediatric Oncology, Hacettepe University, Sihhiye, 06100, Ankara, Turkey
| | - Mustafa Tezer Kutluk
- Faculty of Medicine, Department of Pediatric Oncology, Hacettepe University, Sihhiye, 06100, Ankara, Turkey
| | - Gokcen Coban Cifci
- Faculty of Medicine, Department of Radiology, Hacettepe University, Sihhiye, 06100, Ankara, Turkey
| | - Gozde Yazici
- Faculty of Medicine, Department of Radiation Oncology, Hacettepe University, Sihhiye, 06100, Ankara, Turkey.
| |
Collapse
|
11
|
Yan M, Wang B, Han B, Li Z, Liu X, Liu P. Insights From H3-Wildtype Diffuse Midline Glioma With EZHIP Overexpression. Am J Surg Pathol 2025:00000478-990000000-00490. [PMID: 40098385 DOI: 10.1097/pas.0000000000002388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Affiliation(s)
- Minjun Yan
- Department of Neurosurgery Beijing Tiantan Hospital Capital Medical University
| | - Bo Wang
- Department of Neurosurgery Beijing Tiantan Hospital Capital Medical University
| | - Bo Han
- Department of Neurosurgery Beijing Tiantan Hospital Capital Medical University
| | - Zhuo Li
- Department of Neuropathology, Beijing Neurosurgical Institute Capital Medical University, Beijing, China
| | - Xing Liu
- Department of Neuropathology, Beijing Neurosurgical Institute Capital Medical University, Beijing, China
| | - Pinan Liu
- Department of Neurosurgery Beijing Tiantan Hospital Capital Medical University
| |
Collapse
|
12
|
Kiran S, Xue Y, Sarker DB, Sang QXA. Effects of Induced Pluripotent Stem Cell-Derived Astrocytes on Cisplatin Sensitivity in Pediatric Brain Cancer Cells. Cancers (Basel) 2025; 17:997. [PMID: 40149331 PMCID: PMC11940393 DOI: 10.3390/cancers17060997] [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: 01/27/2025] [Revised: 03/06/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
Background: ATRTs and DIPGs are deadly pediatric brain tumors with poor prognosis. These tumors can develop resistance to chemotherapies, which may be significantly influenced by their microenvironment. Since astrocytes are the most abundant glial cell type in the brain microenvironment and may support tumor growth and chemoresistance, this study investigated the effects of induced pluripotent stem cell-derived astrocytes (iPSC-astrocytes) on cisplatin sensitivity in CHLA-05-ATRT and SF8628 (DIPG) cells. iPSCs provide an unlimited and standardized source of nascent astrocytes, which enables modeling the interaction between childhood brain tumor cells and iPSC-astrocytes within a controlled coculture system. Methods: To study the effects on tumor growth, the iPSC-astrocytes were cocultured with tumor cells. Additionally, the tumor cells were exposed to various concentrations of cisplatin to evaluate their chemosensitivity in the presence of astrocytes. Results: The paracrine interaction of iPSC-astrocytes with tumor cells upregulated astrocyte activation markers GFAP and STAT3 and promoted tumor cell proliferation. Moreover, the cisplatin treatment significantly decreased the viability of CHLA-05-ATRT and SF8628 cells. However, tumor cells exhibited reduced sensitivity to cisplatin in the coculture with iPSC-astrocytes. During cisplatin treatment, DIPG cells in particular showed upregulation of resistance markers, ERK1, STAT3, and MTDH, which are associated with enhanced proliferation and invasion. They also had increased expression of APEX1, which is involved in the base excision repair pathway following cisplatin-induced DNA damage. Conclusion: These findings underscore the significance of the tumor microenvironment in modulating tumor cell survival and chemosensitivity.
Collapse
Affiliation(s)
- Sonia Kiran
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA; (S.K.); (Y.X.); (D.B.S.)
| | - Yu Xue
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA; (S.K.); (Y.X.); (D.B.S.)
| | - Drishty B. Sarker
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA; (S.K.); (Y.X.); (D.B.S.)
| | - Qing-Xiang Amy Sang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA; (S.K.); (Y.X.); (D.B.S.)
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
13
|
Poorva P, Mast J, Cao B, Shah MV, Pollok KE, Shen J. Killing the killers: Natural killer cell therapy targeting glioma stem cells in high-grade glioma. Mol Ther 2025:S1525-0016(25)00168-6. [PMID: 40040281 DOI: 10.1016/j.ymthe.2025.02.043] [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: 10/28/2024] [Revised: 01/22/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025] Open
Abstract
High-grade gliomas (HGGs), including glioblastoma (GBM) in adults and diffuse intrinsic pontine glioma (DIPG) in children, are among the most aggressive and deadly brain tumors. A key factor in their resilience is the presence of glioma stem cells (GSCs), which drive tumor initiation, progression, and resistance to treatment. Targeting and eradicating GSCs holds potential for curing both GBM and DIPG. Natural killer (NK) cells, as part of the innate immune system, naturally recognize and destroy malignant cells. Recent advances in NK cell-based therapies, such as chimeric antigen receptor (CAR)-NK cells, NK cell engagers, and NK cell-derived exosomes, offer promising approaches for treating GBM and DIPG, particularly by addressing the persistence of GSCs. This review highlights these advancements, explores challenges such as the blood-brain barrier and the immunosuppressive tumor microenvironment, and proposes future directions for improving and clinically advancing these NK cell-based therapies for HGGs.
Collapse
Affiliation(s)
- Poorva Poorva
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA
| | - Jensen Mast
- Biochemistry Graduate Program, Indiana University, Bloomington, IN 47405, USA
| | - Bihui Cao
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA
| | - Mitesh V Shah
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Karen E Pollok
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | - Jia Shen
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
14
|
Fukuda Y, Endo M, Ogawa K, Takahashi S, Nakamura M, Kawahara M, Akahane K, Mori H, Gomi A, Shirai K. Long-Term Outcomes of Radiation Therapy for Pediatric Brain Tumors: A Single-Center Study. Cureus 2025; 17:e81282. [PMID: 40291172 PMCID: PMC12032858 DOI: 10.7759/cureus.81282] [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] [Accepted: 03/26/2025] [Indexed: 04/30/2025] Open
Abstract
Brain tumors are the leading cause of mortality among pediatric patients. Recent advancements in genetic analysis have facilitated the development of new therapeutic agents, and high-precision radiotherapy techniques have improved survival rates for certain pediatric brain tumors. However, owing to the rarity of these tumors and the diversity of histological types, most treatment results are reported in clinical trials, and real-world data on the long-term treatment effects of radiotherapy in Japan are scarce. This study investigated the long-term outcomes of pediatric brain tumor treatment at a single institution. A total of 54 pediatric brain tumor patients aged ≤14 years who had undergone radiotherapy between 2007 and 2021 were included. Irradiation was performed using three-dimensional conformal or intensity-modulated radiation therapy. The distribution of each tumor type was as follows: eight diffuse intrinsic pontine gliomas, six malignant gliomas, 12 medulloblastomas, eight ependymomas, 15 germ cell tumors, and five other tumors (malignant peripheral nerve sheath tumor, pinealoblastoma, atypical teratoma/rhabdoid tumor, primitive neuroectoderm tumor, and malignant astroblastoma). The median follow-up duration for all patients and survivors was 48.4 months and 110 months, respectively. The one-, five-, and 10-year overall survival rates according to tumor type were as follows: diffuse intrinsic pontine glioma - 12.5%, 0%, and 0%; malignant glioma - 50%, 0%, and 0%; medulloblastoma - 91.7%, 83.3%, and 58.3%; ependymoma - 100%, 50%, and 37.5%; germ cell tumors - 93.3%, 93.3%, and 93.3%; and others - 80%, 80%, and 40%, respectively. The one-, five-, and 10-year progression-free survival rates according to the tumor type were 0% for both diffuse intrinsic pontine gliomas and malignant gliomas; 75%, 50%, and 50% for medulloblastoma; 62.5%, 25%, and not available for ependymoma; 86.7%, 80%, and 80% for germ cell tumor; and 60%, 40%, and not available for other tumors, respectively. Adverse events of grade 3 or higher (based on common terminology criteria for adverse events version 5.0) were observed in three patients as follows: two with hearing impairment and one with secondary cancer. Our findings revealed that the prognosis and recurrence patterns such as local and disseminated recurrence substantially differ depending on the tumor type. This confirms that each tumor type requires a unique approach. In recent years, significant progress has been made in the stratification and optimization of treatment through genetic analysis. However, to achieve improved tumor control and minimize late effects, the accumulation of long-term clinical data is essential.
Collapse
Affiliation(s)
- Yukiko Fukuda
- Department of Radiology, Jichi Medical University Hospital, Tochigi, JPN
- Department of Radiology, Jichi Medical University Saitama Medical Center, Saitama, JPN
| | - Masashi Endo
- Department of Radiology, Jichi Medical University Hospital, Tochigi, JPN
| | - Kazunari Ogawa
- Department of Radiology, Jichi Medical University Hospital, Tochigi, JPN
| | - Satoru Takahashi
- Department of Radiology, Jichi Medical University Hospital, Tochigi, JPN
| | - Michiko Nakamura
- Department of Radiology, Jichi Medical University Hospital, Tochigi, JPN
| | - Masahiro Kawahara
- Department of Radiology, Jichi Medical University Saitama Medical Center, Saitama, JPN
| | - Keiko Akahane
- Department of Radiology, Jichi Medical University Saitama Medical Center, Saitama, JPN
| | - Harushi Mori
- Department of Radiology, Jichi Medical University Hospital, Tochigi, JPN
| | - Akira Gomi
- Department of Pediatric Neurosurgery, Jichi Children's Medical Center Tochigi, Tochigi, JPN
| | - Katsuyuki Shirai
- Department of Radiology, Jichi Medical University Hospital, Tochigi, JPN
- Department of Radiology, Jichi Medical University Saitama Medical Center, Saitama, JPN
| |
Collapse
|
15
|
Souweidane MM, Bander ED, Zanzonico P, Reiner AS, Manino N, Haque S, Carrasquillo JA, Lyashchenko SK, Thakur SB, Lewis JS, Donzelli M, Cheung NKV, Larson SM, Kramer K, Pandit-Taskar N, Dunkel IJ. Phase 1 dose-escalation trial using convection-enhanced delivery (CED) of radio-immunotheranostic 124I-Omburtamab for diffuse intrinsic pontine glioma (DIPG). Neuro Oncol 2025:noaf039. [PMID: 39969230 DOI: 10.1093/neuonc/noaf039] [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: 10/30/2024] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Median survival for patients with Diffuse Intrinsic Pontine Glioma (DIPG) is 8-12 months. METHODS A phase 1, open label, 3 + 3 dose escalation trial delivered radiolabeled 124I-Omburtamab, targeting B7-H3, using MR-guided stereotactic convection enhanced delivery (CED) into the brainstem of pediatric DIPG patients. CED was performed after completion of standard-of-care external-beam radiation therapy (EBRT). Fifty children were treated and evaluable. 124I-Omburtamab activity was escalated from 0.25-10.0 mCi (9.25-370 MBq) and volume escalated from 0.25 ml-10.0 ml with serial PET/MRI post-administration. Safety was the primary outcome. National Cancer Institute Common Terminology Criteria for Adverse Events were assessed for 30 days following CED of 124I-Omburtamab. Secondary outcomes included overall survival and lesion-to-whole-body absorbed dose ratio. RESULTS The maximum tolerated activity per study protocol was determined to be 6mCi (222 MBq). The overall mean (±SD) total absorbed dose in the lesion per unit injected activity was 35.2 ± 18 cGy/MBq with a high lesion-to-whole-body absorbed dose ratio averaging 816, across all activity levels. Eleven patients had treatment-related grade 3 CNS toxicities with no grade-4 or -5 CNS toxicities. Five dose-limiting toxicity events occurred. Median survival was 15.29 months from diagnosis (95% CI: 12.20 - 16.83 months). Survival rate estimates at 1, 2, and 3 years were 65.4% (CI 53.3-80.1%), 18.4% (CI: 10.2-33.2%), and 11.7% (CI: 5.3-25.7%), respectively. CONCLUSIONS Administration of 124I-Omburtamab via CED is a safe treatment option for DIPG, with a maximum tolerated activity level identified. This study represents the first in-human theranostic use of a 124I radiopharmaceutical, simultaneously, as an imaging and therapeutic agent.
Collapse
Affiliation(s)
- Mark M Souweidane
- Department of Neurological Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
- Department of Neurological Surgery, New York-Presbyterian Hospital/Weill Cornell Medicine, New York, NY 10065, USA
| | - Evan D Bander
- Department of Neurological Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
- Department of Neurological Surgery, New York-Presbyterian Hospital/Weill Cornell Medicine, New York, NY 10065, USA
| | - Pat Zanzonico
- Department of Nuclear Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Anne S Reiner
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Nicole Manino
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Sofia Haque
- Department of Neuroradiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Jorge A Carrasquillo
- Department of Nuclear Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Serge K Lyashchenko
- Department of Radiochemisty, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Sunitha B Thakur
- Department of Medical Physics and Radiology; Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Jason S Lewis
- Department of Radiochemisty, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Maria Donzelli
- Department of Neurological Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Steven M Larson
- Department of Nuclear Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Kim Kramer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Neeta Pandit-Taskar
- Department of Nuclear Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Ira J Dunkel
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
16
|
Rechberger JS, Toll SA, Biswas S, You HB, Chow WD, Kendall N, Navalkele P, Khatua S. Advances in the Repurposing and Blood-Brain Barrier Penetrance of Drugs in Pediatric Brain Tumors. Cancers (Basel) 2025; 17:439. [PMID: 39941807 PMCID: PMC11816256 DOI: 10.3390/cancers17030439] [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: 12/02/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Central nervous system (CNS) tumors are the leading cause of cancer-related mortality in children, with prognosis remaining dismal for some of these malignancies. Though the past two decades have seen advancements in surgery, radiation, and targeted therapy, major unresolved hurdles continue to undermine the therapeutic efficacy. These include challenges in suboptimal drug delivery through the blood-brain barrier (BBB), marked intra-tumoral molecular heterogeneity, and the elusive tumor microenvironment. Drug repurposing or re-tasking FDA-approved drugs with evidence of penetration into the CNS, using newer methods of intracranial drug delivery facilitating optimal drug exposure, has been an area of intense research. This could be a valuable tool, as most of these agents have already gone through the lengthy process of drug development and the evaluation of safety risks and the optimal pharmacokinetic profile. They can now be used and tested in clinics with an accelerated and different approach. Conclusions: The next-generation therapeutic strategy should prioritize repurposing oncologic and non-oncologic drugs that have been used for other indication, and have demonstrated robust preclinical activity against pediatric brain tumors. In combination with novel drug delivery techniques, these drugs could hold significant therapeutic promise in pediatric neurooncology.
Collapse
Affiliation(s)
| | - Stephanie A. Toll
- Children’s Hospital of Michigan, Central Michigan University School of Medicine, Saginaw, MI 48602, USA;
| | - Subhasree Biswas
- Bronglais General Hospital, Caradog Road, Aberystwyth SY23 1ER, Wales, UK;
| | - Hyo Bin You
- Mayo Clinic Alix School of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; (H.B.Y.); (W.D.C.)
| | - William D. Chow
- Mayo Clinic Alix School of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; (H.B.Y.); (W.D.C.)
| | - Nicholas Kendall
- School of Medicine, University of South Dakota Sanford, Vermillion, SD 57069, USA;
| | - Pournima Navalkele
- Division of Oncology, Children’s Hospital of Orange County, Orange, CA 92868, USA;
| | - Soumen Khatua
- Department of Pediatric Hematology/Oncology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| |
Collapse
|
17
|
Madlener S, Stepien N, Senfter D, Mayr L, Laemmerer A, Hedrich C, Baumgartner A, Lötsch-Gojo D, Sterba J, Pokorna P, Kiesel B, Widhalm G, Eckert F, Preusser M, Rössler K, Azizi A, Peyrl A, Czech T, Haberler C, Slavc I, Kasprian G, Dorfer C, Furtner J, Gojo J. Detection of H3F3A K27M or BRAF V600E in liquid biopsies of brain tumor patients as diagnostic and monitoring biomarker: impact of tumor localization and sampling method. Acta Neuropathol 2025; 149:5. [PMID: 39751690 PMCID: PMC11698890 DOI: 10.1007/s00401-024-02842-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/11/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
Gliomas are the most common brain tumor type in children and adolescents. To date, diagnosis and therapy monitoring for these tumors rely on magnetic resonance imaging (MRI) and histopathological as well as molecular analyses of tumor tissue. Recently, liquid biopsies (LB) have emerged as promising tool for diagnosis and longitudinal tumor assessment potentially allowing for a more precise therapeutic management. However, the optimal strategy for monitoring gliomas by LB remains to be determined. In this study, we analyzed circulating tumor DNA (ctDNA) from 78 liquid biopsies (plasma n = 44, cerebrospinal fluid n = 34 (CSF)) of 35 glioma patients, determining H3F3A K28M (K27M) and BRAF V600E mutation allele frequency using droplet digital PCR (ddPCR). All results were correlated to clinically relevant parameters including diagnostic imaging and CSF aspiration site (ventricular vs lumbar) with respect to tumor localization. Regarding diagnostic accuracy, the calculated sensitivity score in the H3F3A K27M cohort was 84.61% for CSF and 73.68% for plasma. In the BRAF V600E cohort, we determined a sensitivity of 83.3% in plasma and 80% in CSF. The overall specificity was 100%. With respect to the CSF aspiration, the intra-operatively obtained CSF demonstrated 100% detection rate, followed by ventricular CSF obtained via Ommaya Reservoir/shunt puncture (93%) and CSF obtained via lumbar puncture (66%). Notably, this further correlated with the proximity of the CSF site to tumor localization. Longitudinal CSF monitoring demonstrated a good correlation to clinical and radiological disease evolution. Importantly, we show for the first time that monitoring BRAF V600E by ddPCR could serve as treatment response assessment in gliomas. In summary, our observation may inform recommendations with regard to location of CSF aspiration when incorporating LB into future treatment protocols.
Collapse
Affiliation(s)
- Sibylle Madlener
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Natalia Stepien
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Daniel Senfter
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Lisa Mayr
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Anna Laemmerer
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Cora Hedrich
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Alicia Baumgartner
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | | | - Jaroslav Sterba
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Petra Pokorna
- Department of Biology, Faculty of Medicine and Central, European Institute of Technology, Masaryk University, Brno, Czech Republic
- Center for Precision Medicine, University Hospital Brno, Brno, Czech Republic
| | - Barbara Kiesel
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Georg Widhalm
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Franziska Eckert
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
| | - Matthias Preusser
- Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Karl Rössler
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Amedeo Azizi
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Andreas Peyrl
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Thomas Czech
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Christine Haberler
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Irene Slavc
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Gregor Kasprian
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Christian Dorfer
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Julia Furtner
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
- Research Center of Image Analysis and Artificial Intelligence (MIAAI), Faculty of Medicine and Dentistry, Danube Private University, Krems-Stein, Austria
| | - Johannes Gojo
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
18
|
Roberts HJ, Ravi K, Marini BL, Schepers A, Kline C, Kilburn L, Prados M, Byron SA, Sturza J, Mueller S, Koschmann C, Franson AT. Retrospective Comparison of Targeted Anticancer Drugs Predicted by the CNS-TAP Tool Versus Those Selected by a Molecularly Driven Tumor Board in Children With DIPG. J Pediatr Hematol Oncol 2025; 47:19-30. [PMID: 39527919 PMCID: PMC11676589 DOI: 10.1097/mph.0000000000002964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/17/2024] [Indexed: 11/16/2024]
Abstract
The recent trial Pediatric Neuro-Oncology Consortium 003 (PNOC003) utilized a molecular tumor board to recommend personalized treatment regimens based on tumor sequencing results in children with DIPG. We separately developed the Central Nervous System Targeted Agent Prediction (CNS-TAP) tool, which numerically scores targeted anticancer agents using preclinical, clinical, and patient-specific data. We hypothesized that highly scored agents from CNS-TAP would overlap with the PNOC003 tumor board's recommendations. For each of the 28 participants, actionable genetic alterations were derived from PNOC003 genomic reports and input to CNS-TAP to identify the highest scoring agents. These agents were then compared with PNOC003 recommendations, with a resultant concordance percentage calculated. Overall, 38% of the total agents recommended by the tumor board were also selected by CNS-TAP, with higher concordance (63%) in a subanalysis including only targeted anticancer agents. Furthermore, nearly all patients (93%) had at least 1 drug chosen by both methods. We demonstrate overlap between agents recommended by CNS-TAP and PNOC003 tumor board, though this does not appear to improve survival. We do observe some discordance, highlighting strengths and limitations of each method. We propose that a combination of expert opinion and data-driven tools may improve targeted treatment recommendations for children with DIPG.
Collapse
Affiliation(s)
| | | | - Bernard L. Marini
- Department of Clinical Pharmacy and Pharmacy Services, University of Michigan
| | - Allison Schepers
- Department of Clinical Pharmacy and Pharmacy Services, University of Michigan
| | - Cassie Kline
- Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | | | | | - Sara A. Byron
- Translational Genomics Research Institute, Phoenix, AZ
| | - Julie Sturza
- University of Michigan Medical School, Ann Arbor, MI
| | - Sabine Mueller
- Neurology, Neurosurgery, and Pediatrics, San Francisco School of Medicine, University of California, San Francisco, CA
- Department of Pediatrics, University of Zurich, Switzerland
| | | | | |
Collapse
|
19
|
King SA, Solst SR, Graham CH, Fiore LZ, Rheem R, Tomanek-Chalkley A, Fath MA, Caster JM, Spitz DR, Howard ME. Additive Effects of Cu-ATSM and Radiation on Survival of Diffuse Intrinsic Pontine Glioma Cells. Radiat Res 2025; 203:10-17. [PMID: 39492578 PMCID: PMC11815956 DOI: 10.1667/rade-24-00076.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024]
Abstract
Diffuse intrinsic pontine gliomas (DIPG) are highly aggressive and treatment-resistant childhood primary brainstem tumors with a median survival of less than one year after diagnosis. The prevailing standard of care for DIPG, radiation therapy, does not prevent fatal disease progression, with most patients succumbing to this disease 3-8 months after completion of radiation therapy. This underscores the urgent need for novel combined-modality approaches for enhancing therapy responses. This study demonstrates that the cellular redox modulating drug, copper (II)-diacetyl-bis(N4-methylthiosemicarbazone) (Cu-ATSM) dose-dependently (1-3 μM) decreased clonogenic cell survival in SU-DIPG50 and SU-DIPG36 cell lines during 6 h of exposure but had no significant effect on survival in normal human astrocytes (NHA). Additional significant (>90%) decreases in DIPG clonogenic survival were observed at 24 h of Cu-ATSM exposure. However, NHAs also began to show dose-dependent 10-70% survival decreases at this point. Notably, 3 μM Cu-ATSM for 6 h resulted in additive clonogenic cell killing of DIPG lines when combined with radiation, which was not seen in NHAs and was partially inhibited by the copper chelator, bathocuproinedisulfonic acid. Cu-ATSM toxicity in DIPG cells was also inhibited by overexpression of mitochondrial-targeted catalase. These results support the hypothesis that Cu-ATSM is selectively cytotoxic to DIPGs by a mechanism involving H2O2 generation and copper and being additively cytotoxic with ionizing radiation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Michelle E. Howard
- Free Radical and Radiation Biology Program, B180 Medical Laboratories, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242
| |
Collapse
|
20
|
Chaturvedi A, Sadashiva N, Kalahasti S, Konar S, Krishna U, Ar P, Shukla D, Beniwal M, Pruthi N, Arimappamagan A, Saini J, Rao S, Santosh V. Pediatric diffuse intrinsic pontine gliomas- a prospective observational study from a tertiary care neurosurgical center. Childs Nerv Syst 2024; 41:71. [PMID: 39730925 DOI: 10.1007/s00381-024-06730-z] [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: 09/26/2024] [Accepted: 12/16/2024] [Indexed: 12/29/2024]
Abstract
INTRODUCTION Diffuse intrinsic pontine glioma (DIPG) in children comprises 80% of brainstem gliomas. In 2021, 5th edition of WHO CNS tumor classification defined H3K27M altered diffuse midline gliomas (DMGs) which replaced this entity. Lesion location precludes resection and the only current option available is radiotherapy. Patient age, duration of symptoms, histone subtype mutation etc. may helpl in prognostication but the disease remains incurable with a median overall survival of 9-12 months. METHOD This is a prospective observational study from a tertiary health care center in a low to middle-income country. We included patients with DIPG (radiological and/or histopathological H3K27M altered) from June 2018 to April 2023. Clinical, radiological, histology, and molecular features were reviewed and prognostic factors for 3 months, 6 months, and overall survival was analysed for all patients. RESULTS We included 92 pediatric patients. The median age of our study population was 8.5 years. Median LPS was 80. Cranial nerve palsy was the most common presenting complaint. Hydrocephalus requiring CSF diversion was present in 38 patients (41.3%). Lesion biopsy was performed in 36 patients (39.1%) and exophytic component decompression was done in 11 patients (11.9%). Seven patients were lost to follow-up. Adjuvant therapy was received by 51 patients (51/85, 60%). Radiotherapy was the only significant prognostic indicator of 3 months, 6 months, and overall survival (HR: 0.39). The presence of necrosis on histopathology was also an indicator of poor prognosis (HR: 2.38). There were 7 long-term survivors in our study but we did not find any significant survival prognostic indicator amongst this group. CONCLUSION Conventional adjuvant therapy has not proven of much benefit. With the advancement in molecular understanding of the entity, there is an upsurge in the development of targeted therapy but with no promising results so far. In this study, we have attempted to explore the prognostic factors and unique challenges we face in a resource-limited setting against this disease.
Collapse
Affiliation(s)
- Aprajita Chaturvedi
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, 560029, India
| | - Nishanth Sadashiva
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, 560029, India.
| | - Sathyarao Kalahasti
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, 560029, India
| | - Subhas Konar
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, 560029, India
| | - Uday Krishna
- Consultant Radiation Oncology, Apollo Proton Cancer Center, Chennai, India
| | - Prabhuraj Ar
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, 560029, India
| | - Dhaval Shukla
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, 560029, India
| | - Manish Beniwal
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, 560029, India
| | - Nupur Pruthi
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, 560029, India
| | - Arivazhagan Arimappamagan
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, 560029, India
| | - Jitender Saini
- Department of Neuroimaging and Interventional Radiology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, 560029, India
| | - Shilpa Rao
- Department Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, 560029, India
| | - Vani Santosh
- Department Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, 560029, India
| |
Collapse
|
21
|
Ronsley R, Bertrand KC, Song EZ, Timpanaro A, Choe M, Tlais D, Vitanza NA, Park JR. CAR T cell therapy for pediatric central nervous system tumors: a review of the literature and current North American trials. Cancer Metastasis Rev 2024; 43:1205-1216. [PMID: 39251462 PMCID: PMC11554695 DOI: 10.1007/s10555-024-10208-4] [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: 04/22/2024] [Accepted: 08/22/2024] [Indexed: 09/11/2024]
Abstract
Central nervous system (CNS) tumors are the leading cause of cancer-related death in children. Typical therapy for CNS tumors in children involves a combination of surgery, radiation, and chemotherapy. While upfront therapy is effective for many high-grade tumors, therapy at the time of relapse remains limited. Furthermore, for diffuse intrinsic pontine glioma (DIPG) and diffuse midline glioma (DMG), there are currently no curative therapies. Chimeric antigen receptor T (CAR T) cell therapy is a promising novel treatment avenue for these tumors. Here, we review the preclinical evidence for CAR T cell use in pediatric brain tumors, the preliminary clinical experience of CNS CAR T cell trials, toxicity associated with systemic and locoregional CAR T cell therapy for CNS tumors, challenges in disease response evaluation with CAR T cell therapy, and the knowledge gained from correlative biologic studies from these trials in the pediatric and young adult population.
Collapse
Affiliation(s)
- Rebecca Ronsley
- Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Kelsey C Bertrand
- Department of Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN, 38105, USA
- Division of Neuro-Oncology, Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Edward Z Song
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Andrea Timpanaro
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Michelle Choe
- Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Dana Tlais
- Department of Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN, 38105, USA
- Division of Neuro-Oncology, Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Nicholas A Vitanza
- Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Julie R Park
- Department of Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN, 38105, USA.
| |
Collapse
|
22
|
Yu X, Li S, Mai W, Hua X, Sun M, Lai M, Zhang D, Xiao Z, Wang L, Shi C, Luo L, Cai L. Pediatric diffuse intrinsic pontine glioma radiotherapy response prediction: MRI morphology and T2 intensity-based quantitative analyses. Eur Radiol 2024; 34:7962-7972. [PMID: 38907098 PMCID: PMC11557687 DOI: 10.1007/s00330-024-10855-9] [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: 12/27/2023] [Revised: 03/31/2024] [Accepted: 04/25/2024] [Indexed: 06/23/2024]
Abstract
OBJECTIVES An easy-to-implement MRI model for predicting partial response (PR) postradiotherapy for diffuse intrinsic pontine glioma (DIPG) is lacking. Utilizing quantitative T2 signal intensity and introducing a visual evaluation method based on T2 signal intensity heterogeneity, and compared MRI radiomic models for predicting radiotherapy response in pediatric patients with DIPG. METHODS We retrospectively included patients with brainstem gliomas aged ≤ 18 years admitted between July 2011 and March 2023. Applying Response Assessment in Pediatric Neuro-Oncology criteria, we categorized patients into PR and non-PR groups. For qualitative analysis, tumor heterogeneity vision was classified into four grades based on T2-weighted images. Quantitative analysis included the relative T2 signal intensity ratio (rT2SR), extra pons volume ratio, and tumor ring-enhancement volume. Radiomic features were extracted from T2-weighted and T1-enhanced images of volumes of interest. Univariate analysis was used to identify independent variables related to PR. Multivariate logistic regression was performed using significant variables (p < 0.05) from univariate analysis. RESULTS Of 140 patients (training n = 109, and test n = 31), 64 (45.7%) achieved PR. The AUC of the predictive model with extrapontine volume ratio, rT2SRmax-min (rT2SRdif), and grade was 0.89. The AUCs of the T2-weighted and T1WI-enhanced models with radiomic signatures were 0.84 and 0.81, respectively. For the 31 DIPG test sets, the AUCs were 0.91, 0.83, and 0.81, for the models incorporating the quantitative features, radiomic model (T2-weighted images, and T1W1-enhanced images), respectively. CONCLUSION Combining T2-weighted quantification with qualitative and extrapontine volume ratios reliably predicted pediatric DIPG radiotherapy response. CLINICAL RELEVANCE STATEMENT Combining T2-weighted quantification with qualitative and extrapontine volume ratios can accurately predict diffuse intrinsic pontine glioma (DIPG) radiotherapy response, which may facilitate personalized treatment and prognostic assessment for patients with DIPG. KEY POINTS Early identification is crucial for radiotherapy response and risk stratification in diffuse intrinsic pontine glioma. The model using tumor heterogeneity and quantitative T2 signal metrics achieved an AUC of 0.91. Using a combination of parameters can effectively predict radiotherapy response in this population.
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
| | - Shaoqun Li
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China
| | - Wenfeng Mai
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Xiaoyu Hua
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Mengnan Sun
- 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
| | - Dong Zhang
- 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
| | - Lichao Wang
- 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.
| | - Linbo Cai
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China.
| |
Collapse
|
23
|
Huchede P, Meyer S, Berthelot C, Hamadou M, Bertrand-Chapel A, Rakotomalala A, Manceau L, Tomine J, Lespinasse N, Lewandowski P, Cordier-Bussat M, Broutier L, Dutour A, Rochet I, Blay JY, Degletagne C, Attignon V, Montero-Carcaboso A, Le Grand M, Pasquier E, Vasiljevic A, Gilardi-Hebenstreit P, Meignan S, Leblond P, Ribes V, Cosset E, Castets M. BMP2 and BMP7 cooperate with H3.3K27M to promote quiescence and invasiveness in pediatric diffuse midline gliomas. eLife 2024; 12:RP91313. [PMID: 39373720 PMCID: PMC11458179 DOI: 10.7554/elife.91313] [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] [Indexed: 10/08/2024] Open
Abstract
Pediatric diffuse midline gliomas (pDMG) are an aggressive type of childhood cancer with a fatal outcome. Their major epigenetic determinism has become clear, notably with the identification of K27M mutations in histone H3. However, the synergistic oncogenic mechanisms that induce and maintain tumor cell phenotype have yet to be deciphered. In 20 to 30% of cases, these tumors have an altered BMP signaling pathway with an oncogenic mutation on the BMP type I receptor ALK2, encoded by ACVR1. However, the potential impact of the BMP pathway in tumors non-mutated for ACVR1 is less clear. By integrating bulk, single-cell, and spatial transcriptomic data, we show here that the BMP signaling pathway is activated at similar levels between ACVR1 wild-type and mutant tumors and identify BMP2 and BMP7 as putative activators of the pathway in a specific subpopulation of cells. By using both pediatric isogenic glioma lines genetically modified to overexpress H3.3K27M and patients-derived DIPG cell lines, we demonstrate that BMP2/7 synergizes with H3.3K27M to induce a transcriptomic rewiring associated with a quiescent but invasive cell state. These data suggest a generic oncogenic role for the BMP pathway in gliomagenesis of pDMG and pave the way for specific targeting of downstream effectors mediating the K27M/BMP crosstalk.
Collapse
Affiliation(s)
- Paul Huchede
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Swann Meyer
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Clement Berthelot
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Maud Hamadou
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Adrien Bertrand-Chapel
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Andria Rakotomalala
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER Cancer Heterogeneity Plasticity and Resistance to Therapies, Centre Oscar LambretLilleFrance
| | - Line Manceau
- Université Paris Cité, CNRS, Institut Jacques MonodParisFrance
| | - Julia Tomine
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Nicolas Lespinasse
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Paul Lewandowski
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER Cancer Heterogeneity Plasticity and Resistance to Therapies, Centre Oscar LambretLilleFrance
| | - Martine Cordier-Bussat
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Laura Broutier
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Aurelie Dutour
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Isabelle Rochet
- Multisite Institute of Pathology, Groupement Hospitalier Est du CHU de Lyon, Hôpital Femme-Mère EnfantBronFrance
| | - Jean-Yves Blay
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | | | | | - Angel Montero-Carcaboso
- Preclinical Therapeutics and Drug Delivery Research Program, Department of Oncology, Hospital Sant Joan de DéuBarcelonaSpain
| | - Marion Le Grand
- Centre de Recherche en Cancérologie de Marseille (CRCM), Université Aix-Marseille, Institut Paoli- Calmettes, Centre de Lutte Contre le Cancer de la région PACA, INSERM 1068, CNRS 7258MarseilleFrance
| | - Eddy Pasquier
- Centre de Recherche en Cancérologie de Marseille (CRCM), Université Aix-Marseille, Institut Paoli- Calmettes, Centre de Lutte Contre le Cancer de la région PACA, INSERM 1068, CNRS 7258MarseilleFrance
| | - Alexandre Vasiljevic
- Multisite Institute of Pathology, Groupement Hospitalier Est du CHU de Lyon, Hôpital Femme-Mère EnfantBronFrance
| | | | - Samuel Meignan
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER Cancer Heterogeneity Plasticity and Resistance to Therapies, Centre Oscar LambretLilleFrance
| | - Pierre Leblond
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
- Department of Pediatric Oncology, Institute of Pediatric Hematology and Oncology (IHOPe), Centre Léon BérardLyonFrance
| | - Vanessa Ribes
- Université Paris Cité, CNRS, Institut Jacques MonodParisFrance
| | - Erika Cosset
- GLIMMER Of lIght (GLIoblastoma MetabolisM, HetERogeneity, and OrganoIds) team, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| | - Marie Castets
- Childhood Cancer & Cell Death (C3) team, LabEx DEVweCAN, Institut Convergence Plascan, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286LyonFrance
| |
Collapse
|
24
|
Bayramova E, Manklow L, Filyridou M, Pilat A. Ophthalmic Presentation of Diffuse Intrinsic Pontine Glioma in Children (Case Series and Literature Review). Neuroophthalmology 2024; 49:200-205. [PMID: 40190377 PMCID: PMC11970730 DOI: 10.1080/01658107.2024.2402726] [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: 12/02/2022] [Revised: 07/29/2024] [Accepted: 09/06/2024] [Indexed: 04/09/2025] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a rare aggressive brainstem lesion, affecting mainly young children. This report describes sixth cranial nerve palsy as the initial ophthalmic presentation in children with this pathology. Case series and literature review. All children presented with sixth nerve palsy were consecutively recruited from the pediatric clinic at the East Sussex NHS Healthcare Trust within the last 10 years. Full ophthalmic examination, orthoptic assessment, and refraction check were done in three patients. Magnetic Resonance Imaging was carried out using 1.5 Tesla (Siemens Symphony, Erlangen, Germany) to establish the diagnosis. The patients' age ranged from 5 to 14 years at the time of presentation. All presented with sudden onset esotropia and limited abduction, suggestive of presence of sixth nerve palsy, requiring urgent medical attention. On detailed questioning and assessment, all children showed various neurological symptoms including nystagmus, liquid dysphagia, balance problems, and nocturnal enuresis. Two out of three patients died within 7 months following diagnosis. Sudden onset esotropia, especially due to sixth nerve palsy in children, should be considered a red flag symptom, prompting proper urgent specialist assessment. Sixth nerve palsy in patients with DIPG was associated with severely reduced life expectancy in this case series of three patients, shorter than in reported non-ophthalmic presentations.
Collapse
Affiliation(s)
| | - Lorraine Manklow
- Department of Ophthalmology, East Sussex NHS Healthcare Trust, Eastbourne, UK
| | - Maria Filyridou
- Department of Ophthalmology, East Sussex NHS Healthcare Trust, Eastbourne, UK
| | - Anastasia Pilat
- Department of Ophthalmology, East Sussex NHS Healthcare Trust, Eastbourne, UK
- Ulverscroft Unit, Ulverscroft Eye Unit, University of Leicester, Leicester, UK
| |
Collapse
|
25
|
Chaturvedi A, Sadashiva N, Shukla D, Konar S, Krishna U, Beniwal M, Rao S, Santosh V, Saini J, Vazhayil V, Prabhuraj AR, Pruthi N, Arimappamagan A. Thalamic H3K27M altered diffuse midline gliomas: Clinicopathological and outcome analysis. Clin Neurol Neurosurg 2024; 244:108449. [PMID: 39053322 DOI: 10.1016/j.clineuro.2024.108449] [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: 05/16/2024] [Revised: 07/03/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024]
Abstract
INTRODUCTION Diffuse midline glioma (DMG) is a relatively new entity which was introduced in the fourth edition of the WHO classification of CNS tumours in 2016 and later underwent revision in 2021. It is an infiltrative glioma arising from midline structures, viz., thalamus, spine, and brainstem. Current literature on DMG is based majorly on brainstem lesions, and DMGs arising elsewhere remain unexplored. In our study, we have discussed our experience with thalamic DMGs. METHODOLOGY This is a retrospective observational study of all patients with histopathologically proven DMG H3K27M altered, arising in the thalamus from 2018 to 2022. Clinical, neuroimaging, and pathology were re-reviewed, and prognostic factors for 3 months, 6 months, and overall survival (OS) were analyzed for all patients. RESULTS There were 89 patients- 64 adults and 25 pediatric patients with thalamic DMG. The median age at presentation was 24 years. Raised ICP followed by limb weakness were the most common presenting complaints. Stereotactic biopsy was performed in 64 (71.9 %) patients and surgical decompression in 25 (28.1 %) patients. CSF diversion was required in 53 (59.6 %) patients. Median survival was 8 months in adults and 7 months in pediatric (p-value: 0.51). Raised ICP and TP53 mutation were prognostic factors in pediatric population. Radiotherapy with or without chemotherapy improved survival (p-value- <0.01). CONCLUSION Thalamic DMGs have a poor prognosis which is comparable to brainstem DMGs. Radiotherapy improves survival in these patients. However, the disease remains an enigma and further work delving into its molecular characterization should be encouraged.
Collapse
Affiliation(s)
- Aprajita Chaturvedi
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Nishanth Sadashiva
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bangalore 560029, India.
| | - Dhaval Shukla
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Subhas Konar
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Uday Krishna
- Consultant Radiation Oncology, Apollo Proton Cancer Center, Chennai, India
| | - Manish Beniwal
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Shilpa Rao
- Department Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Vani Santosh
- Department Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Jitender Saini
- Department of Neuroimaging and Interventional Radiology, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Vikas Vazhayil
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - A R Prabhuraj
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Nupur Pruthi
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Arivazhagan Arimappamagan
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| |
Collapse
|
26
|
Guidara S, Seyve A, Poncet D, Leonce C, Bringuier PP, McLeer A, Sturm D, Cartalat S, Picart T, Ferrari A, Hench J, Frank S, Meyronet D, Ducray F, Barritault M. Characteristics of H3K27M-mutant diffuse gliomas with a non-midline location. J Neurooncol 2024; 169:391-398. [PMID: 38937309 DOI: 10.1007/s11060-024-04733-z] [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: 05/02/2024] [Accepted: 05/31/2024] [Indexed: 06/29/2024]
Abstract
PURPOSE Diffuse midline gliomas (DMG) with H3K27 alterations (H3K27M-DMG) are a highly aggressive form of brain cancer. In rare cases, H3K27 mutations have been observed in diffuse non-midline gliomas (DNMG). It is currently unclear how these tumors should be classified. Herein, we analyze the characteristics of DNMG with H3K27M mutations. METHODS We reviewed the clinical, radiological and histological characteristics of all patients with an H3K27M mutated diffuse glioma diagnosed in our institution, between 2016 and 2023, to identify cases with a non-midline location. We then performed a molecular characterization (DNA methylation profiling, whole genome and transcriptome sequencing or targeted sequencing) of patients with an H3K27M-mutant DNMG and reviewed previously reported cases. RESULTS Among 51 patients (18 children and 33 adults) diagnosed with an H3K27M diffuse glioma, we identified two patients (4%) who had a non-midline location. Including our two patients, 39 patients were reported in the literature with an H3K27M-mutant DNMG. Tumors were most frequently located in the temporal lobe (48%), affected adolescents and adults, and were associated with a poor outcome (median overall survival was 10.3 months (0.1-84)). Median age at diagnosis was 19.1 years. Tumors frequently harbored TP53 mutations (74%), ATRX mutations (71%) and PDGFRA mutations or amplifications (44%). In DNA methylation analysis, H3K27M-mutant DNMG clustered within or close to the reference group of H3K27M-mutant DMG. Compared to their midline counterpart, non-midline gliomas with H3K27M mutations seemed more frequently associated with PDGFRA alterations. CONCLUSION DNMG with H3K27M mutations share many similarities with their midline counterpart, suggesting that they correspond to a rare anatomical presentation of these tumors. This is of paramount importance, as they may benefit from new therapeutic approaches such as ONC201.
Collapse
Affiliation(s)
- Souhir Guidara
- Department of Medical Genetics, Hedi Chaker Hospital, Sfax, Tunisia.
| | - Antoine Seyve
- Department of Neurooncology, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France
| | - Delphine Poncet
- Department of Pathology, Groupe Hospitalier Est, Hospices Civils de Lyon, Bron, France
- Institut Neuro Myo Gène (INMG), Pathophysiology and Genetics of Neuron and Muscle (PGNM), Université Claude Bernard Lyon 1, CNRS UMR 5261-INSERM U1315, Neuron-Muscle interaction team, Lyon, France
| | - Camille Leonce
- Department of Pathology, Groupe Hospitalier Est, Hospices Civils de Lyon, Bron, France
- Centre Léon Bérard, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Cancer Research Center of Lyon, Lyon, France
| | - Pierre-Paul Bringuier
- Department of Pathology, Groupe Hospitalier Est, Hospices Civils de Lyon, Bron, France
- Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Anne McLeer
- Service d'Anatomie et Cytologie Pathologiques CHU Grenoble Alpes, Institute for Advanced Biosciences UGA, Université Grenoble Alpes, INSERM U1209/CNRS 5309, Grenoble, France
| | - Dominik Sturm
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Pediatric Glioma Research Group, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stéphanie Cartalat
- Department of Neurooncology, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France
| | - Thiebaud Picart
- Centre Léon Bérard, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Cancer Research Center of Lyon, Lyon, France
- Université Claude Bernard Lyon 1, Villeurbanne, France
- Department of Neurosurgery, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France
| | - Anthony Ferrari
- Gilles Thomas Bioinformatics Platform, Synergie Lyon Cancer, Cancer Research Center of Lyon, Centre Léon Bérard FR, Lyon, France
| | - Jürgen Hench
- Institute for Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Stephan Frank
- Institute for Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - David Meyronet
- Department of Pathology, Groupe Hospitalier Est, Hospices Civils de Lyon, Bron, France
- Université Claude Bernard Lyon 1, Villeurbanne, France
- Cancer Initiation and Tumoral Cell Identity Department, Cancer Research Centre of Lyon (CRCL) INSERM 1052, CNRS 5286, Lyon, France
| | - François Ducray
- Department of Neurooncology, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France
- Université Claude Bernard Lyon 1, Villeurbanne, France
- Cancer Initiation and Tumoral Cell Identity Department, Cancer Research Centre of Lyon (CRCL) INSERM 1052, CNRS 5286, Lyon, France
| | - Marc Barritault
- Department of Pathology, Groupe Hospitalier Est, Hospices Civils de Lyon, Bron, France.
- Cancer Initiation and Tumoral Cell Identity Department, Cancer Research Centre of Lyon (CRCL) INSERM 1052, CNRS 5286, Lyon, France.
| |
Collapse
|
27
|
Wawrzuta D, Chojnacka M, Drogosiewicz M, Pędziwiatr K, Dembowska-Bagińska B. Reirradiation for diffuse intrinsic pontine glioma: prognostic radiomic factors at progression. Strahlenther Onkol 2024; 200:797-804. [PMID: 38748214 PMCID: PMC11343881 DOI: 10.1007/s00066-024-02241-7] [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: 12/19/2023] [Accepted: 04/23/2024] [Indexed: 08/24/2024]
Abstract
PURPOSE Diffuse intrinsic pontine glioma (DIPG) is a lethal pediatric brain tumor. Radiation therapy (RT) is the standard treatment, with reirradiation considered in case of progression. However, the prognostic factors for reirradiation are not well understood. This study aims to investigate the outcomes of DIPG patients undergoing reirradiation and identify clinical and radiomic prognostic factors. METHODS We conducted a retrospective analysis of patients with DIPG who underwent reirradiation at our institution between January 2016 and December 2023. Using PyRadiomics, we extracted radiomic features of tumors at the time of progression from FLAIR MRI images and collected clinical data. We used the least absolute shrinkage and selection operator (lasso) for Cox's proportional hazard model with leave-one-out cross-validation to select optimal prognostic factors for survival after reirradiation. RESULTS The study included 18 patients who underwent reirradiation at first progression, receiving a total dose of 20 Gy or 24 Gy in 2‑Gy fractions. Reirradiation was well tolerated, with no severe toxicity. Most patients (78%) showed neurological improvement after treatment. Median survival after progression was 29.2 weeks. The Cox model demonstrated a concordance of 0.81 (95% CI: 0.75-0.88), revealing that tumor sphericity and structural gray-level heterogeneity in FLAIR MRI images were associated with longer survival of reirradiated patients. CONCLUSION Reirradiation is a safe and effective approach for patients with DIPG. MRI-based radiomic models could be helpful in predicting survival after reirradiation.
Collapse
Affiliation(s)
- Dominik Wawrzuta
- Department of Radiation Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Wawelska 15B, 02-034, Warsaw, Poland.
| | - Marzanna Chojnacka
- Department of Radiation Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Wawelska 15B, 02-034, Warsaw, Poland
| | - Monika Drogosiewicz
- Department of Oncology, Children's Memorial Health Institute, Al. Dzieci Polskich 20, 04-730, Warsaw, Poland
| | - Katarzyna Pędziwiatr
- Department of Radiation Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Wawelska 15B, 02-034, Warsaw, Poland
| | - Bożenna Dembowska-Bagińska
- Department of Oncology, Children's Memorial Health Institute, Al. Dzieci Polskich 20, 04-730, Warsaw, Poland
| |
Collapse
|
28
|
Akdemir EY, Odia Y, Hall MD, Mehta MP, Kotecha R. An Update on H3K27M-altered Diffuse Midline Glioma: Diagnostic and Therapeutic Challenges in Clinical Practice. Pract Radiat Oncol 2024; 14:443-451. [PMID: 38704025 DOI: 10.1016/j.prro.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024]
Abstract
H3K27-altered diffuse midline glioma (DMG H3K27-altered) is a relatively newly-designated WHO entity which primarily affects the midline structures of the central nervous system (CNS), including the brainstem (predominantly pontine region), thalamus, midbrain, or spinal cord, and primarily affects children and young adults. Despite the proximity of these tumors to eloquent areas in the CNS, novel stereotactic approaches have facilitated the ability to obtain tissue diagnoses without significant morbidity, providing molecular diagnostic information in more than half of patients. Conventionally fractionated radiation therapy to a total dose of 54-60 Gy in 27-30 fractions and 24 Gy in 12 fractions play a crucial role in the definitive treatment of these tumors in the primary and salvage settings, respectively. Hypofractionated regimens may allow for accelerated treatment courses in selected patients without jeopardizing disease control or survival. The decision to add concurrent or adjuvant systemic therapy mainly relies on the physicians' experience without solid evidence in the literature in favor of any particular regimen. Recently, novel agents, such as ONC201 have demonstrated promising oncologic outcomes in progressive/recurrent tumors and are currently under investigation in ongoing randomized trials. Given the scarcity of data and well-established guidelines due to the rare nature of the disease, we provide a contemporary overview on the molecular underpinnings of this disease entity, describe the role of radiotherapy and systemic therapy, and present practice management principles based on the published literature.
Collapse
Affiliation(s)
- Eyub Yasar Akdemir
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida
| | - Yazmin Odia
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida; Department of Neuro-Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida
| | - Matthew D Hall
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida; Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Minesh P Mehta
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida; Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Rupesh Kotecha
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida; Herbert Wertheim College of Medicine, Florida International University, Miami, Florida.
| |
Collapse
|
29
|
Lin FY, Stuckert A, Tat C, White M, Ruggieri L, Zhang H, Mehta B, Lapteva N, Mei Z, Major A, Thakkar S, Shum T, Parikh K, Wu MF, Lindsay HB, Scherer L, Shekar M, Baxter P, Wang T, Grilley B, Moeller K, Hicks J, Roy A, Anastas J, Malbari F, Aldave G, Chintagumpala M, Blaney S, Parsons DW, Brenner MK, Heslop HE, Rooney CM, Omer B. Phase I Trial of GD2.CART Cells Augmented With Constitutive Interleukin-7 Receptor for Treatment of High-Grade Pediatric CNS Tumors. J Clin Oncol 2024; 42:2769-2779. [PMID: 38771986 PMCID: PMC11305939 DOI: 10.1200/jco.23.02019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/26/2023] [Accepted: 02/12/2024] [Indexed: 05/23/2024] Open
Abstract
PURPOSE T cells modified with chimeric antigen receptors (CARTs) have demonstrated efficacy for hematologic malignancies; however, benefit for patients with CNS tumors has been limited. To enhance T cell activity against GD2+ CNS malignancies, we modified GD2-directed CART cells (GD2.CARTs) with a constitutively active interleukin (IL)-7 receptor (C7R-GD2.CARTs). METHODS Patients age 1-21 years with H3K27-altered diffuse midline glioma (DMG) or other recurrent GD2-expressing CNS tumors were eligible for this phase I trial (ClinicalTrials.gov identifier: NCT04099797). All subjects received standard-of-care adjuvant radiation therapy or chemotherapy before study enrollment. The first treatment cohort received GD2.CARTs alone (1 × 107 cells/m2), and subsequent cohorts received C7R-GD2.CARTs at two dose levels (1 × 107 cells/m2; 3 × 107 cells/m2). Standard lymphodepletion with cyclophosphamide and fludarabine was included at all dose levels. RESULTS Eleven patients (age 4-18 years) received therapy without dose-limiting toxicity. The GD2.CART cohort did not experience toxicity, but had disease progression after brief improvement of residual neurologic deficits (≤3 weeks). The C7R-GD2.CART cohort developed grade 1 tumor inflammation-associated neurotoxicity in seven of eight (88%) cases, controllable with anakinra. Cytokine release syndrome was observed in six of eight (75%, grade 1 in all but one patient) and associated with increased circulating IL-6 and IP-10 (P < .05). Patients receiving C7R-GD2.CARTs experienced temporary improvement from baseline neurologic deficits (range, 2 to >12 months), and seven of eight (88%) remained eligible for additional treatment cycles (range 2-4 cycles). Partial responses by iRANO criteria were observed in two of seven (29%) patients with DMG treated by C7R-GD2.CARTs. CONCLUSION Intravenous GD2.CARTs with and without C7R were well tolerated. Patients treated with C7R-GD2.CARTs exhibited transient improvement of neurologic deficits and increased circulating cytokines/chemokines. Treatment with C7R-GD2.CARTs represents a novel approach warranting further investigation for children with these incurable CNS cancers.
Collapse
Affiliation(s)
- Frank Y. Lin
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX
- Dan L Duncan Comprehensive Cancer Center, Houston, TX
| | - Austin Stuckert
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX
| | - Candise Tat
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX
| | - Mark White
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX
| | - Lucia Ruggieri
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX
| | - Huimin Zhang
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Birju Mehta
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Natalia Lapteva
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Zhuyong Mei
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Angela Major
- Department of Pathology, Baylor College of Medicine, Houston, TX
| | - Sachin Thakkar
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Thomas Shum
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
- Department of Radiology, Brigham and Women's Hospital, Boston, MA
| | - Kathan Parikh
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Meng-Fen Wu
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Holly B. Lindsay
- Department of Pediatrics Heme-Onc and Bone Marrow Transplantation, Children's Hospital Colorado Center for Cancer and Blood Disorders, University of Colorado Anschutz Medical Campus, Denver, CO
| | - Lauren Scherer
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX
| | - Meghan Shekar
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX
| | - Patricia Baxter
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX
- Dan L Duncan Comprehensive Cancer Center, Houston, TX
| | - Tao Wang
- Dan L Duncan Comprehensive Cancer Center, Houston, TX
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Bambi Grilley
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Karen Moeller
- Department of Radiology, Baylor College of Medicine, Houston, TX
| | - John Hicks
- Department of Pathology, Baylor College of Medicine, Houston, TX
| | - Angshumoy Roy
- Dan L Duncan Comprehensive Cancer Center, Houston, TX
- Department of Pathology, Baylor College of Medicine, Houston, TX
| | - Jamie Anastas
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX
| | - Fatema Malbari
- Department of Neurology, Baylor College of Medicine, Houston, TX
| | - Guillermo Aldave
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX
| | - Murali Chintagumpala
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX
- Dan L Duncan Comprehensive Cancer Center, Houston, TX
| | - Susan Blaney
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX
- Dan L Duncan Comprehensive Cancer Center, Houston, TX
| | - D. Williams Parsons
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX
- Dan L Duncan Comprehensive Cancer Center, Houston, TX
| | - Malcolm K. Brenner
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX
- Dan L Duncan Comprehensive Cancer Center, Houston, TX
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Helen E. Heslop
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX
- Dan L Duncan Comprehensive Cancer Center, Houston, TX
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Cliona M. Rooney
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX
- Dan L Duncan Comprehensive Cancer Center, Houston, TX
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Bilal Omer
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX
- Dan L Duncan Comprehensive Cancer Center, Houston, TX
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| |
Collapse
|
30
|
Bonada M, Pittarello M, De Fazio E, Gans A, Alimonti P, Slika H, Legnani F, Di Meco F, Tyler B. Pediatric Hemispheric High-Grade Gliomas and H3.3-G34 Mutation: A Review of the Literature on Biological Features and New Therapeutic Strategies. Genes (Basel) 2024; 15:1038. [PMID: 39202398 PMCID: PMC11353413 DOI: 10.3390/genes15081038] [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: 06/20/2024] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 09/03/2024] Open
Abstract
Pediatric high-grade glioma (pHGG) encompasses a wide range of gliomas with different genomic, epigenomic, and transcriptomic features. Almost 50% of pHGGs present a mutation in genes coding for histone 3, including the subtype harboring the H3.3-G34 mutation. In this context, histone mutations are frequently associated with mutations in TP53 and ATRX, along with PDGFRA and NOTCH2NL amplifications. Moreover, the H3.3-G34 histone mutation induces epigenetic changes in immune-related genes and exerts modulatory functions on the microenvironment. Also, the functionality of the blood-brain barrier (BBB) has an impact on treatment response. The prognosis remains poor with conventional treatments, thus eliciting the investigation of additional and alternative therapies. Promising molecular targets include PDGFRA amplification, BRAF mutation, EGFR amplification, NF1 loss, and IDH mutation. Considering that pHGGs harboring the H3.3-G34R mutation appear to be more susceptible to immunotherapies (ITs), different options have been recently explored, including immune checkpoint inhibitors, antibody mediated IT, and Car-T cells. This review aims to summarize the knowledge concerning cancer biology and cancer-immune cell interaction in this set of pediatric gliomas, with a focus on possible therapeutic options.
Collapse
Affiliation(s)
- Marta Bonada
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (M.B.); (F.L.); (F.D.M.)
- Department of Oncology and Hemato-Oncology, University of Milan School of Medicine, Via Rudini 8, 20122 Milan, Italy;
| | - Matilde Pittarello
- Department of Biomedical Sciences, Humanitas University, 20072 Milan, Italy;
| | - Emerson De Fazio
- Department of Medicine, Vita-Salute San Raffaele University School of Medicine, 20132 Milan, Italy;
| | - Alessandro Gans
- Department of Oncology and Hemato-Oncology, University of Milan School of Medicine, Via Rudini 8, 20122 Milan, Italy;
- ASST Ovest Milanese, Neurology and Stroke Unit, Neuroscience Department, 20025 Legnano, Italy
| | - Paolo Alimonti
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02120, USA;
| | - Hasan Slika
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
| | - Federico Legnani
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (M.B.); (F.L.); (F.D.M.)
- Department of Oncology and Hemato-Oncology, University of Milan School of Medicine, Via Rudini 8, 20122 Milan, Italy;
| | - Francesco Di Meco
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (M.B.); (F.L.); (F.D.M.)
- Department of Oncology and Hemato-Oncology, University of Milan School of Medicine, Via Rudini 8, 20122 Milan, Italy;
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
| | - Betty Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
| |
Collapse
|
31
|
de la Nava D, Ausejo-Mauleon I, Laspidea V, Gonzalez-Huarriz M, Lacalle A, Casares N, Zalacain M, Marrodan L, García-Moure M, Ochoa MC, Tallon-Cobos AC, Hernandez-Osuna R, Marco-Sanz J, Dhandapani L, Hervás-Corpión I, Becher OJ, Nazarian J, Mueller S, Phoenix TN, van der Lugt J, Hernaez M, Guruceaga E, Koschmann C, Venneti S, Allen JE, Dun MD, Fueyo J, Gomez-Manzano C, Gallego Perez-Larraya J, Patiño-García A, Labiano S, Alonso MM. The oncolytic adenovirus Delta-24-RGD in combination with ONC201 induces a potent antitumor response in pediatric high-grade and diffuse midline glioma models. Neuro Oncol 2024; 26:1509-1525. [PMID: 38554031 PMCID: PMC11300018 DOI: 10.1093/neuonc/noae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Indexed: 04/01/2024] Open
Abstract
BACKGROUND Pediatric high-grade gliomas (pHGGs), including diffuse midline gliomas (DMGs), are aggressive pediatric tumors with one of the poorest prognoses. Delta-24-RGD and ONC201 have shown promising efficacy as single agents for these tumors. However, the combination of both agents has not been evaluated. METHODS The production of functional viruses was assessed by immunoblotting and replication assays. The antitumor effect was evaluated in a panel of human and murine pHGG and DMG cell lines. RNAseq, the seahorse stress test, mitochondrial DNA content, and γH2A.X immunofluorescence were used to perform mechanistic studies. Mouse models of both diseases were used to assess the efficacy of the combination in vivo. The tumor immune microenvironment was evaluated using flow cytometry, RNAseq, and multiplexed immunofluorescence staining. RESULTS The Delta-24-RGD/ONC201 combination did not affect the virus replication capability in human pHGG and DMG models in vitro. Cytotoxicity analysis showed that the combination treatment was either synergistic or additive. Mechanistically, the combination treatment increased nuclear DNA damage and maintained the metabolic perturbation and mitochondrial damage caused by each agent alone. Delta-24-RGD/ONC201 cotreatment extended the overall survival of mice implanted with human and murine pHGG and DMG cells, independent of H3 mutation status and location. Finally, combination treatment in murine DMG models revealed a reshaping of the tumor microenvironment to a proinflammatory phenotype. CONCLUSIONS The Delta-24-RGD/ONC201 combination improved the efficacy compared to each agent alone in in vitro and in vivo models by potentiating nuclear DNA damage and in turn improving the antitumor (immune) response to each agent alone.
Collapse
Affiliation(s)
- Daniel de la Nava
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Iker Ausejo-Mauleon
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Virginia Laspidea
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Marisol Gonzalez-Huarriz
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Andrea Lacalle
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Noelia Casares
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Marta Zalacain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Lucía Marrodan
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Marc García-Moure
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Maria C Ochoa
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Antonio Carlos Tallon-Cobos
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Reyes Hernandez-Osuna
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Javier Marco-Sanz
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Laasya Dhandapani
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Irati Hervás-Corpión
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Oren J Becher
- Jack Martin Fund Division of Pediatric Hematology-oncology, Mount Sinai, New York, USA
| | - Javad Nazarian
- Division of Oncology and Children’s Research Center, DIPG/DMG Research Center Zurich, University Children’s Hospital Zurich, Zurich, Switzerland
- Virginia Tech University, Washington, District of Columbia, USA
- Children’s National Health System, Center for Genetic Medicine Research, Washington, District of Columbia, USA
| | - Sabine Mueller
- University of California, San FranciscoSan Francisco, California, USA
- Division of Oncology and Children’s Research Center, DIPG/DMG Research Center Zurich, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Timothy N Phoenix
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio, USA
| | | | - Mikel Hernaez
- Bioinformatics Platform, Center for Applied Medical Research, University of Navarra (CIMA), Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Elizabeth Guruceaga
- Bioinformatics Platform, Center for Applied Medical Research, University of Navarra (CIMA), Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Carl Koschmann
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | - Sriram Venneti
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Matthew D Dun
- Paediatric Stream, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine, and Wellbeing, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
| | - Juan Fueyo
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Candelaria Gomez-Manzano
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jaime Gallego Perez-Larraya
- Department of Neurology, Clínica Universidad de Navarra, Pamplona, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Ana Patiño-García
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Sara Labiano
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| | - Marta M Alonso
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Spain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
32
|
Blasco-Santana L, Colmenero I. Molecular and Pathological Features of Paediatric High-Grade Gliomas. Int J Mol Sci 2024; 25:8498. [PMID: 39126064 PMCID: PMC11312892 DOI: 10.3390/ijms25158498] [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: 05/28/2024] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Paediatric high-grade gliomas are among the most common malignancies found in children. Despite morphological similarities to their adult counterparts, there are profound biological and molecular differences. Furthermore, and thanks to molecular biology, the diagnostic pathology of paediatric high-grade gliomas has experimented a dramatic shift towards molecular classification, with important prognostic implications, as is appropriately reflected in both the current WHO Classification of Tumours of the Central Nervous System and the WHO Classification of Paediatric Tumours. Emphasis is placed on histone 3, IDH1, and IDH2 alterations, and on Receptor of Tyrosine Kinase fusions. In this review we present the current diagnostic categories from the diagnostic pathology perspective including molecular features.
Collapse
Affiliation(s)
- Luis Blasco-Santana
- Pathology Department, Hospital Infantil Universitario del Niño Jesús, Avenida de Menéndez Pelayo, 65, 28009 Madrid, Spain
| | - Isabel Colmenero
- Pathology Department, Hospital Infantil Universitario del Niño Jesús, Avenida de Menéndez Pelayo, 65, 28009 Madrid, Spain
| |
Collapse
|
33
|
Bautista F, Verdú-Amorós J, Geoerger B, Rubio-San-Simón A, Paoletti X, Zwaan CM, Casanova M, Marshall LV, Carceller F, Doz F, Lecinse C, Vassal G, Pearson ADJ, Kearns P, Moreno L. Evolution of the Innovative Therapies for Children With Cancer Consortium Trial Portfolio for Drug Development for Children With Cancer. J Clin Oncol 2024; 42:2516-2526. [PMID: 38743911 DOI: 10.1200/jco.23.01237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 02/09/2024] [Accepted: 02/29/2024] [Indexed: 05/16/2024] Open
Abstract
PURPOSE The aim of the Innovative Therapies for Children with Cancer (ITCC) consortium is to improve access to novel therapies for children and adolescents with cancer. The evolution of the ITCC clinical trial portfolio since 2003 was reviewed. METHODS All ITCC-labeled phase I/II trials opened between January 1, 2003 and February 3, 2018 were analyzed in two periods (2003-2010 and 2011-2018), and data were extracted from the ITCC database, regulatory agencies' registries, and publications. RESULTS Sixty-one trials (62% industry-sponsored) enrolled 3,198 patients. The number of trials in the second period increased by almost 300% (16 v 45). All biomarker-driven trials (n = 14) were conducted in the second period. The use of rolling six and model-based designs increased (1 of 9, 11% v 21 of 31, 68%), and that of 3 + 3 designs decreased (5 of 9, 55% v 5 of 31, 16%; P = .014). The proportion of studies evaluating chemotherapeutics only decreased (5 of 16, 31% v 4 of 45, 9%), the proportion of single-agent targeted therapies did not change (9 of 16, 56.2% v 24 of 45, 53.3%), the proportion of combination targeted therapies trials increased (2 of 16, 12%, v 17 of 45, 38%), the proportion of randomized phase II trials increased (1 of 7, 14% v 8 of 14, 57%). More trials were part of a pediatric investigation plan in the second period (4 of 16, 25% v 21 of 45, 46%). The median time for Ethics Committees' approvals was 1.7 times longer for academic compared with industry-sponsored trials. CONCLUSION This study reports a shift in the paradigm of early drug development for childhood cancers, with more biologically relevant targets evaluated in biomarker-driven trials or in combination with other therapies and with more model-based or randomized designs and a greater focus on fulfilling regulatory requirements. Improvement of trial setup and recruitment could increase the number of patients benefiting from novel agents.
Collapse
Affiliation(s)
- Francisco Bautista
- Division of Pediatric Hematology and Oncology, Hospital Universitario Niño Jesús, Madrid, Spain
- Princess Máxima Center, Utrecht, the Netherlands
| | - Jaime Verdú-Amorós
- Division of Pediatric Hematology and Oncology, Hospital Universitario Niño Jesús, Madrid, Spain
- Division of Pediatric Hematology and Oncology, Hospital Clínico Universitario de Valencia, Biomedical Research Institute, INCLIVA, Valencia, Spain
| | - Birgit Geoerger
- Pediatric and Adolescent Oncology Department, Gustave Roussy Cancer Campus, INSERM U1015, Université Paris-Saclay, Villejuif, France
| | - Alba Rubio-San-Simón
- Division of Pediatric Hematology and Oncology, Hospital Universitario Niño Jesús, Madrid, Spain
| | - Xavier Paoletti
- Institut Curie & Université Versailles St Quentin & INSERM U900 STAMPM, Paris, France
| | - C Michel Zwaan
- Princess Máxima Center, Utrecht, the Netherlands
- Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Michela Casanova
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Lynley V Marshall
- Pediatric and Adolescent Oncology Drug Development, Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Fernando Carceller
- Pediatric and Adolescent Oncology Drug Development, Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Francois Doz
- SIREDO Cancer Center (Care, Innovation and Research in Pediatric, Adolescents, and Young Adults Oncology), Curie Institute Paris, and University Paris Cité, Paris, France
| | - Carole Lecinse
- Innovative Therapies for Children with Cancer, Gustave Roussy Cancer Campus, Villejuif, France
| | - Gilles Vassal
- Innovative Therapies for Children with Cancer, Gustave Roussy Cancer Campus, Villejuif, France
| | - Andrew D J Pearson
- Pediatric and Adolescent Oncology Drug Development, Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Pamela Kearns
- Institute of Cancer and Genomic Sciences, NIHR Birmingham Biomedical Research Centre, University of Birmingham, Birmingham, United Kingdom
| | - Lucas Moreno
- Division of Pediatric Hematology and Oncology, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| |
Collapse
|
34
|
Boretti A. Improving chimeric antigen receptor T-cell therapies by using artificial intelligence and internet of things technologies: A narrative review. Eur J Pharmacol 2024; 974:176618. [PMID: 38679117 DOI: 10.1016/j.ejphar.2024.176618] [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: 01/20/2024] [Revised: 04/18/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024]
Abstract
Cancer poses a formidable challenge in the field of medical science, prompting the exploration of innovative and efficient treatment strategies. One revolutionary breakthrough in cancer therapy is Chimeric Antigen Receptor (CAR) T-cell therapy, an avant-garde method involving the customization of a patient's immune cells to combat cancer. Particularly successful in addressing blood cancers, CAR T-cell therapy introduces an unprecedented level of effectiveness, offering the prospect of sustained disease management. As ongoing research advances to overcome current challenges, CAR T-cell therapy stands poised to become an essential tool in the fight against cancer. Ongoing enhancements aim to improve its effectiveness and reduce time and cost, with the integration of Artificial Intelligence (AI) and Internet of Things (IoT) technologies. The synergy of AI and IoT could enable more precise tailoring of CAR T-cell therapy to individual patients, streamlining the therapeutic process. This holds the potential to elevate treatment efficacy, mitigate adverse effects, and expedite the overall progress of CAR T-cell therapies.
Collapse
Affiliation(s)
- Alberto Boretti
- Independent Scientist, Johnsonville, Wellington, New Zealand.
| |
Collapse
|
35
|
Sepulveda F, Scotto Opipari R, Coppola F, Ramaglia A, Mankad K, Alves CAP, Bison B, Löbel U. Approaches to supratentorial brain tumours in children. Neuroradiology 2024:10.1007/s00234-024-03398-9. [PMID: 38953989 DOI: 10.1007/s00234-024-03398-9] [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: 04/02/2024] [Accepted: 06/01/2024] [Indexed: 07/04/2024]
Abstract
The differential diagnosis of supratentorial brain tumours in children can be challenging, especially considering the recent changes to the WHO classification of CNS tumours published in 2021. Many new tumour types have been proposed which frequently present in children and young adults and their imaging features are currently being described by the neuroradiology community. The purpose of this article is to provide guidance to residents and fellows new to the field of paediatric neuroradiology on how to evaluate an MRI of a patient with a newly diagnosed supratentorial tumour. Six different approaches are discussed including: 1. Tumour types, briefly discussing the main changes to the recent WHO classification of CNS tumours, 2. Patient age and its influence on incidence rates of specific tumour types, 3. Growth patterns, 4. Tumour location and how defining the correct location helps in narrowing down the differential diagnoses and 5. Imaging features of the tumour on DWI, SWI, FLAIR and post contrast sequences.
Collapse
Affiliation(s)
- Francisco Sepulveda
- Departamento de Imagenología, Clínica Alemana de Santiago, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | | | - Fiorenza Coppola
- Department of Diagnostic and Interventional Radiology, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| | - Antonia Ramaglia
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Kshitij Mankad
- Department of Radiology, Great Ormond Street Hospital for Children, London, UK
| | - Cesar A P Alves
- Radiology Department, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Brigitte Bison
- Diagnostic and Interventional Neuroradiology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Ulrike Löbel
- Department of Radiology, Great Ormond Street Hospital for Children, London, UK.
| |
Collapse
|
36
|
Morin L, Godard F, Aubriot-Lorton MH, Drouet C. Whole Spinal Cord FDG Uptake at PET/CT Caused by H3K27-Altered Diffuse Midline Glioma in an Adult Patient. Clin Nucl Med 2024; 49:e321-e323. [PMID: 38775827 DOI: 10.1097/rlu.0000000000005246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
ABSTRACT Diffuse midline glioma, H3K27-altered, is a relatively new entity, characterized by H3K27M histone mutation. This rare pediatric disease with severe prognosis has recently been identified as a new subtype of diffuse astrocytoma due to major breakthrough in histopathological and molecular characterization of gliomas. We report a case of H3K27-altered diffuse midline glioma in a 30-year-old woman.
Collapse
Affiliation(s)
- Léo Morin
- From the Pathology Department, University Hospital François Mitterrand
| | - François Godard
- Nuclear Medicine Department, Centre Georges François Leclerc, Dijon, France
| | | | - Clément Drouet
- Nuclear Medicine Department, Centre Georges François Leclerc, Dijon, France
| |
Collapse
|
37
|
Nonnenbroich LF, Bouchal SM, Millesi E, Rechberger JS, Khatua S, Daniels DJ. H3K27-Altered Diffuse Midline Glioma of the Brainstem: From Molecular Mechanisms to Targeted Interventions. Cells 2024; 13:1122. [PMID: 38994974 PMCID: PMC11240752 DOI: 10.3390/cells13131122] [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: 06/03/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/13/2024] Open
Abstract
Pediatric high-grade gliomas are a devastating subset of brain tumors, characterized by their aggressive pathophysiology and limited treatment options. Among them, H3 K27-altered diffuse midline gliomas (DMG) of the brainstem stand out due to their distinct molecular features and dismal prognosis. Recent advances in molecular profiling techniques have unveiled the critical role of H3 K27 alterations, particularly a lysine-to-methionine mutation on position 27 (K27M) of the histone H3 tail, in the pathogenesis of DMG. These mutations result in epigenetic dysregulation, which leads to altered chromatin structure and gene expression patterns in DMG tumor cells, ultimately contributing to the aggressive phenotype of DMG. The exploration of targeted therapeutic avenues for DMG has gained momentum in recent years. Therapies, including epigenetic modifiers, kinase inhibitors, and immunotherapies, are under active investigation; these approaches aim to disrupt aberrant signaling cascades and overcome the various mechanisms of therapeutic resistance in DMG. Challenges, including blood-brain barrier penetration and DMG tumor heterogeneity, require innovative approaches to improve drug delivery and personalized treatment strategies. This review aims to provide a comprehensive overview of the evolving understanding of DMG, focusing on the intricate molecular mechanisms driving tumorigenesis/tumor progression and the current landscape of emerging targeted interventions.
Collapse
Affiliation(s)
- Leo F. Nonnenbroich
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (L.F.N.); (J.S.R.)
- Hopp Children’s Cancer Center, Heidelberg (KiTZ), 69120 Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
| | - Samantha M. Bouchal
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (L.F.N.); (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Elena Millesi
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Research Laboratory of the Division of Plastic and Reconstructive Surgery, Department of Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Julian S. Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (L.F.N.); (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Soumen Khatua
- Department of Pediatric Hematology/Oncology, Section of Neuro-Oncology, Mayo Clinic, Rochester, MN 55905, USA;
| | - David J. Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (L.F.N.); (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
38
|
Gue R, Lakhani DA. The 2021 World Health Organization Central Nervous System Tumor Classification: The Spectrum of Diffuse Gliomas. Biomedicines 2024; 12:1349. [PMID: 38927556 PMCID: PMC11202067 DOI: 10.3390/biomedicines12061349] [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: 05/13/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
The 2021 edition of the World Health Organization (WHO) classification of central nervous system tumors introduces significant revisions across various tumor types. These updates, encompassing changes in diagnostic techniques, genomic integration, terminology, and grading, are crucial for radiologists, who play a critical role in interpreting brain tumor imaging. Such changes impact the diagnosis and management of nearly all central nervous system tumor categories, including the reclassification, addition, and removal of specific tumor entities. Given their pivotal role in patient care, radiologists must remain conversant with these revisions to effectively contribute to multidisciplinary tumor boards and collaborate with peers in neuro-oncology, neurosurgery, radiation oncology, and neuropathology. This knowledge is essential not only for accurate diagnosis and staging, but also for understanding the molecular and genetic underpinnings of tumors, which can influence treatment decisions and prognostication. This review, therefore, focuses on the most pertinent updates concerning the classification of adult diffuse gliomas, highlighting the aspects most relevant to radiological practice. Emphasis is placed on the implications of new genetic information on tumor behavior and imaging findings, providing necessary tools to stay abreast of advancements in the field. This comprehensive overview aims to enhance the radiologist's ability to integrate new WHO classification criteria into everyday practice, ultimately improving patient outcomes through informed and precise imaging assessments.
Collapse
Affiliation(s)
- Racine Gue
- Department of Neuroradiology, West Virginia University, Morgantown, WV 26506, USA
| | - Dhairya A. Lakhani
- Department of Neuroradiology, West Virginia University, Morgantown, WV 26506, USA
- Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
39
|
Zhou T, Qiao B, Peng B, Liu Y, Gong Z, Kang M, He Y, Pang C, Dai Y, Sheng M. Predicting histological grade in pediatric glioma using multiparametric radiomics and conventional MRI features. Sci Rep 2024; 14:13683. [PMID: 38871755 PMCID: PMC11176337 DOI: 10.1038/s41598-024-63222-5] [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: 11/15/2023] [Accepted: 05/27/2024] [Indexed: 06/15/2024] Open
Abstract
Prediction of glioma is crucial to provide a precise treatment plan to optimize the prognosis of children with glioma. However, studies on the grading of pediatric gliomas using radiomics are limited. Meanwhile, existing methods are mainly based on only radiomics features, ignoring intuitive information about tumor morphology on traditional imaging features. This study aims to utilize multiparametric magnetic resonance imaging (MRI) to identify high-grade and low-grade gliomas in children and establish a classification model based on radiomics features and clinical features. A total of 85 children with gliomas underwent tumor resection, and part of the tumor tissue was examined pathologically. Patients were categorized into high-grade and low-grade groups according to World Health Organization guidelines. Preoperative multiparametric MRI data, including contrast-enhanced T1-weighted imaging, T2-weighted imaging, T2-weighted fluid-attenuated inversion recovery, diffusion-weighted images, and apparent diffusion coefficient sequences, were obtained and labeled by two radiologists. The images were preprocessed, and radiomics features were extracted for each MRI sequence. Feature selection methods were used to select radiomics features, and statistically significant clinical features were identified using t-tests. The selected radiomics features and conventional MRI features were used to train the AutoGluon models. The improved model, based on radiomics features and conventional MRI features, achieved a balanced classification accuracy of 66.59%. The cross-validated areas under the receiver operating characteristic curve for the classifier of AutoGluon frame were 0.8071 on the test dataset. The results indicate that the performance of AutoGluon models can be improved by incorporating conventional MRI features, highlighting the importance of the experience of radiologists in accurately grading pediatric gliomas. This method can help predict the grade of pediatric glioma before pathological examination and assist in determining the appropriate treatment plan, including radiotherapy, chemotherapy, drugs, and gene surgery.
Collapse
Affiliation(s)
- Tengfei Zhou
- Department of Radiology, Children's Hospital of Soochow University, Suzhou, China
| | - Baobao Qiao
- School of Life Science and Technology, Changchun University of Science and Technology, Changchun, China
| | - Bo Peng
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
- Jinan Guoke Medical Engineering Technology Development Co., Ltd, Jinan, China
| | - Yuqi Liu
- Department of Radiology, Children's Hospital of Soochow University, Suzhou, China
| | - Zhenjia Gong
- School of Life Science and Technology, Changchun University of Science and Technology, Changchun, China
| | - Mengfei Kang
- Department of Radiology, Children's Hospital of Soochow University, Suzhou, China
| | - Yu He
- Department of Radiology, Children's Hospital of Soochow University, Suzhou, China
| | - Chunying Pang
- School of Life Science and Technology, Changchun University of Science and Technology, Changchun, China
| | - Yakang Dai
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China.
- Jinan Guoke Medical Engineering Technology Development Co., Ltd, Jinan, China.
| | - Mao Sheng
- Department of Radiology, Children's Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
40
|
Valvi S, Manoharan N, Mateos MK, Hassall TE, Ziegler DS, McCowage GB, Dun MD, Eisenstat DD, Gottardo NG, Hansford JR. Management of patients with diffuse intrinsic pontine glioma in Australia and New Zealand: Australian and New Zealand Children's Haematology/Oncology Group position statement. Med J Aust 2024; 220:533-538. [PMID: 38699949 DOI: 10.5694/mja2.52295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 03/26/2024] [Indexed: 05/05/2024]
Abstract
INTRODUCTION The main mission of the Australian and New Zealand Children's Haematology and Oncology Group (ANZCHOG) is to develop and facilitate local access to the world's leading evidence-based clinical trials for all paediatric cancers, including brain tumours, as soon as practically possible. Diffuse intrinsic pontine gliomas (DIPGs) - a subset of a larger group of tumours now termed diffuse midline glioma, H3K27-altered (DMG) - are paediatric brain cancers with less than 10% survival at two years. In the absence of any proven curative therapies, significant recent advancements have been made in pre-clinical and clinical research, leading many to seek integration of novel therapies early into standard practice. Despite these innovative therapeutic approaches, DIPG remains an incurable disease for which novel surgical, imaging, diagnostic, radiation and systemic therapy approaches are needed. MAIN RECOMMENDATIONS All patients with DIPG should be discussed in multidisciplinary neuro-oncology meetings (including pathologists, neuroradiologists, radiation oncologists, neurosurgeons, medical oncologists) at diagnosis and at relapse or progression. Radiation therapy to the involved field remains the local and international standard of care treatment. Proton therapy does not yield a superior survival outcome compared with photon therapy and patients should undergo radiation therapy with the available modality (photon or proton) at their treatment centre. Patients may receive concurrent chemotherapy or radiation-sensitising agents as part of a clinical trial. Biopsy should be offered to facilitate consideration of experimental therapies and eligibility for clinical trial participation. After radiation therapy, each patient should be managed individually with either observation or considered for enrolment on a clinical trial, if eligible, after full discussion with the family. Re-irradiation can be considered for progressive disease. CHANGES IN MANAGEMENT AS A RESULT OF THE GUIDELINE Every child diagnosed with DIPG should be offered enrolment on a clinical trial where available. Access to investigational drugs without biological rationale outside the clinical trial setting is not supported. In case of potentially actionable target identification with molecular profiling and absence of a suitable clinical trial, rational targeted therapies can be considered through compassionate access programs.
Collapse
Affiliation(s)
- Santosh Valvi
- Perth Children's Hospital, Perth, WA
- Telethon Kids Institute, Perth, WA
- University of Western Australia, Perth, WA
| | - Neevika Manoharan
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, Sydney, NSW
- University of New South Wales, Sydney, NSW
| | - Marion K Mateos
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, Sydney, NSW
- University of New South Wales, Sydney, NSW
| | - Timothy Eg Hassall
- Queensland Children's Hospital, Brisbane, QLD
- Frazer Institute, University of Queensland, Brisbane, QLD
| | - David S Ziegler
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, Sydney, NSW
- University of New South Wales, Sydney, NSW
| | | | - Matthew D Dun
- University of Newcastle, Newcastle, NSW
- Hunter Medical Research Institute, Newcastle, NSW
| | - David D Eisenstat
- Children's Cancer Centre, Royal Children's Hospital Melbourne, Melbourne, VIC
- Murdoch Children's Research Institute, Melbourne, VIC
- University of Melbourne, Melbourne, VIC
| | | | - Jordan R Hansford
- Women's and Children's Hospital, Adelaide, SA
- South Australian Health and Medical Research Institute, Adelaide, SA
- University of Adelaide, Adelaide, SA
| |
Collapse
|
41
|
Özkan A, Yağcı Küpeli B, Küpeli S, Sezgin G, Bayram İ. Nimotuzumab-vinorelbine combination therapy versus other regimens in the treatment of pediatric diffuse intrinsic pontine glioma. Childs Nerv Syst 2024; 40:1671-1680. [PMID: 38478066 DOI: 10.1007/s00381-024-06329-4] [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: 09/05/2023] [Accepted: 02/21/2024] [Indexed: 05/23/2024]
Abstract
PURPOSE Pediatric diffuse intrinsic pontine glioma (DIPG) is a fatal disease associated with a median survival of < 1 year despite aggressive treatments. This retrospective study analyzed the treatment outcomes of patients aged < 18 years who were diagnosed with DIPG between 2012 and 2022 and who received different chemotherapy regimens. METHODS After radiotherapy, patients with DIPG received nimotuzumab-vinorelbine combination or temozolomide-containing therapy. When nimotuzumab was unavailable, it was replaced by vincristine, etoposide, and carboplatin/cyclophosphamide (VECC). Temozolomide was administered as a single agent or a part of the combination chemotherapy comprising temozolomide, irinotecan, and bevacizumab. Furthermore, 1- and 3-year overall survival (OS), progression-free survival (PFS), and median OS and PFS were analyzed. RESULTS The median age of 40 patients with DIPG was 97 ± 46.93 (23-213) months; the median follow-up time was 12 months. One and 3-year OS were 35.0% and 7.5%, respectively. Median OS was 12 months in all patients (n = 40), and it was 16, 10, and 11 months in those who received first-line nimotuzumab-vinorelbine combination (n = 13), temozolomide-based (n = 14), and VECC (n = 6) chemotherapy regimens, respectively (p = 0.360). One patient who received gefitinib survived for 16 months. Conversely, patients who never received radiotherapy and any antineoplastic medicamentous therapy (n = 6) had a median OS of 4 months. CONCLUSION Nimotuzumab-vinorelbine combination therapy prolonged OS by 6 months compared with temozolomide-containing chemotherapy, although the difference was not statistically significant.
Collapse
Affiliation(s)
- Ayşe Özkan
- Department of Pediatric Oncology and Pediatric Bone Marrow Transplantation Unit, Faculty of Medicine, Balcali Hospital, Çukurova University, Adana, Turkey.
| | - Begül Yağcı Küpeli
- Department of Pediatric Hematology and Oncology, Adana City Training and Research Hospital, University of Health Sciences, Adana, Turkey
| | - Serhan Küpeli
- Department of Pediatric Oncology and Pediatric Bone Marrow Transplantation Unit, Faculty of Medicine, Balcali Hospital, Çukurova University, Adana, Turkey
| | - Gülay Sezgin
- Department of Pediatric Oncology and Pediatric Bone Marrow Transplantation Unit, Faculty of Medicine, Balcali Hospital, Çukurova University, Adana, Turkey
| | - İbrahim Bayram
- Department of Pediatric Oncology and Pediatric Bone Marrow Transplantation Unit, Faculty of Medicine, Balcali Hospital, Çukurova University, Adana, Turkey
| |
Collapse
|
42
|
Arms LM, Duchatel RJ, Jackson ER, Sobrinho PG, Dun MD, Hua S. Current status and advances to improving drug delivery in diffuse intrinsic pontine glioma. J Control Release 2024; 370:835-865. [PMID: 38744345 DOI: 10.1016/j.jconrel.2024.05.018] [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: 12/05/2023] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024]
Abstract
Diffuse midline glioma (DMG), including tumors diagnosed in the brainstem (diffuse intrinsic pontine glioma - DIPG), is the primary cause of brain tumor-related death in pediatric patients. DIPG is characterized by a median survival of <12 months from diagnosis, harboring the worst 5-year survival rate of any cancer. Corticosteroids and radiation are the mainstay of therapy; however, they only provide transient relief from the devastating neurological symptoms. Numerous therapies have been investigated for DIPG, but the majority have been unsuccessful in demonstrating a survival benefit beyond radiation alone. Although many barriers hinder brain drug delivery in DIPG, one of the most significant challenges is the blood-brain barrier (BBB). Therapeutic compounds must possess specific properties to enable efficient passage across the BBB. In brain cancer, the BBB is referred to as the blood-brain tumor barrier (BBTB), where tumors disrupt the structure and function of the BBB, which may provide opportunities for drug delivery. However, the biological characteristics of the brainstem's BBB/BBTB, both under normal physiological conditions and in response to DIPG, are poorly understood, which further complicates treatment. Better characterization of the changes that occur in the BBB/BBTB of DIPG patients is essential, as this informs future treatment strategies. Many novel drug delivery technologies have been investigated to bypass or disrupt the BBB/BBTB, including convection enhanced delivery, focused ultrasound, nanoparticle-mediated delivery, and intranasal delivery, all of which are yet to be clinically established for the treatment of DIPG. Herein, we review what is known about the BBB/BBTB and discuss the current status, limitations, and advances of conventional and novel treatments to improving brain drug delivery in DIPG.
Collapse
Affiliation(s)
- Lauren M Arms
- Therapeutic Targeting Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Paediatric Program, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine & Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - Ryan J Duchatel
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Paediatric Program, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine & Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - Evangeline R Jackson
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Paediatric Program, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine & Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - Pedro Garcia Sobrinho
- Therapeutic Targeting Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Matthew D Dun
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Paediatric Program, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine & Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - Susan Hua
- Therapeutic Targeting Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Paediatric Program, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine & Wellbeing, University of Newcastle, Callaghan, NSW, Australia.
| |
Collapse
|
43
|
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
|
44
|
Dun MD, Odia Y, Arrillaga-Romany I. Diffuse midline glioma, H3K27-altered: Illuminating the dark side of the moon. Neuro Oncol 2024; 26:S89-S91. [PMID: 38108088 PMCID: PMC11066917 DOI: 10.1093/neuonc/noad245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Indexed: 12/19/2023] Open
Affiliation(s)
- Matthew D Dun
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Paediatric Stream, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
| | - Yazmin Odia
- Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | | |
Collapse
|
45
|
Odia Y, Hall MD, Cloughesy TF, Wen PY, Arrillaga-Romany I, Daghistani D, Mehta MP, Tarapore RS, Ramage SC, Allen JE. Selective DRD2 antagonist and ClpP agonist ONC201 in a recurrent non-midline H3 K27M-mutant glioma cohort. Neuro Oncol 2024; 26:S165-S172. [PMID: 38386699 PMCID: PMC11066928 DOI: 10.1093/neuonc/noae021] [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: 07/10/2023] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND Diffuse midline glioma, H3 K27-altered (H3 K27M-altered DMG) are invariably lethal, disproportionately affecting the young and without effective treatment besides radiotherapy. The 2016 World Health Organization (WHO) Central Nervous System (CNS) Tumors Classification defined H3 K27M mutations as pathognomonic but restricted diagnosis to diffuse gliomas involving midline structures by 2018. Dordaviprone (ONC201) is an oral investigational small molecule, DRD2 antagonist, and ClpP agonist associated with durable responses in recurrent H3 K27M-mutant DMG. Activity of ONC201 in non-midline H3 K27M-mutant diffuse gliomas has not been reported. METHODS Patients with recurrent non-midline H3 K27M-mutant diffuse gliomas treated with ONC201 were enrolled in 5 trials. Eligibility included measurable disease by Response Assessment in Neuro-Oncology (RANO) high-grade glioma, Karnofsky/Lansky performance score ≥60, and ≥90 days from radiation. The primary endpoint was overall response rate (ORR). RESULTS Five patients with cerebral gliomas (3 frontal, 1 temporal, and 1 parietal) met inclusion. One complete and one partial response were reported by investigators. Blinded independent central review confirmed ORR by RANO criteria for 2, however, 1 deemed nonmeasurable and another stable. A responding patient also noted improved mobility and alertness. CONCLUSIONS H3 K27M-mutant diffuse gliomas occasionally occur in non-midline cerebrum. ONC201 exhibits activity in H3 K27M-mutant gliomas irrespective of CNS location.
Collapse
Affiliation(s)
- Yazmin Odia
- Department of Neuro-Oncology, Miami Cancer Institute (MCI), Baptist Health South Florida, Miami, Florida, USA
| | - Matthew D Hall
- Department of Neuro-Oncology, Miami Cancer Institute (MCI), Baptist Health South Florida, Miami, Florida, USA
| | | | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women’s Cancer Center (DFCC), Harvard Medical School, Boston, Massachusetts, USA
| | - Isabel Arrillaga-Romany
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women’s Cancer Center (DFCC), Harvard Medical School, Boston, Massachusetts, USA
| | - Doured Daghistani
- Department of Neuro-Oncology, Miami Cancer Institute (MCI), Baptist Health South Florida, Miami, Florida, USA
| | - Minesh P Mehta
- Department of Neuro-Oncology, Miami Cancer Institute (MCI), Baptist Health South Florida, Miami, Florida, USA
| | | | | | | |
Collapse
|
46
|
Jackson ER, Persson ML, Fish CJ, Findlay IJ, Mueller S, Nazarian J, Hulleman E, van der Lugt J, Duchatel RJ, Dun MD. A review of current therapeutics targeting the mitochondrial protease ClpP in diffuse midline glioma, H3 K27-altered. Neuro Oncol 2024; 26:S136-S154. [PMID: 37589388 PMCID: PMC11066926 DOI: 10.1093/neuonc/noad144] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Indexed: 08/18/2023] Open
Abstract
Diffuse midline gliomas (DMGs) are devastating pediatric brain tumors recognized as the leading cause of cancer-related death in children. DMGs are high-grade gliomas (HGGs) diagnosed along the brain's midline. Euchromatin is the hallmark feature of DMG, caused by global hypomethylation of H3K27 either through point mutations in histone H3 genes (H3K27M), or by overexpression of the enhancer of zeste homolog inhibitory protein. In a clinical trial for adults with progressive HGGs, a 22-year-old patient with a thalamic DMG, H3 K27-altered, showed a remarkable clinical and radiological response to dordaviprone (ONC201). This response in an H3 K27-altered HGG patient, coupled with the lack of response of patients harboring wildtype-H3 tumors, has increased the clinical interest in dordaviprone for the treatment of DMG. Additional reports of clinical benefit have emerged, but research defining mechanisms of action (MOA) fall behind dordaviprone's clinical use, with biomarkers of response unresolved. Here, we summarize dordaviprone's safety, interrogate its preclinical MOA identifying the mitochondrial protease "ClpP" as a biomarker of response, and discuss other ClpP agonists, expanding the arsenal of potential weapons in the fight against DMG. Finally, we discuss combination strategies including ClpP agonists, and their immunomodulatory effects suggestive of a role for the tumor microenvironment in DMG patient response.
Collapse
Affiliation(s)
- Evangeline R Jackson
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales , Australia
| | - Mika L Persson
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales , Australia
| | - Cameron J Fish
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales , Australia
| | - Izac J Findlay
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales , Australia
| | - Sabine Mueller
- DIPG/DMG Center Zurich, University Children’s Hospital Zürich, Zurich, Switzerland
- Department of Neurology, Neurosurgery and Pediatric, UCSF, San Francisco, California, USA
| | - Javad Nazarian
- DIPG/DMG Center Zurich, University Children’s Hospital Zürich, Zurich, Switzerland
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, District of Columbia, USA
- The George Washington University, School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Esther Hulleman
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands, Utrecht, Netherlands
| | - Jasper van der Lugt
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands, Utrecht, Netherlands
| | - Ryan J Duchatel
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales , Australia
| | - Matthew D Dun
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales , Australia
- Paediatric Program, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|
47
|
Patel J, Aittaleb R, Doherty R, Gera A, Lau B, Messinger D, Wadden J, Franson A, Saratsis A, Koschmann C. Liquid biopsy in H3K27M diffuse midline glioma. Neuro Oncol 2024; 26:S101-S109. [PMID: 38096156 PMCID: PMC11066927 DOI: 10.1093/neuonc/noad229] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Indexed: 02/15/2024] Open
Abstract
Diffuse midline glioma (DMG) with H3K27M mutation is an aggressive and difficult to treat pediatric brain tumor. Recurrent gain of function mutations in H3.3 (H3.3A) and H3.1 (H3C2) at the 27th lysine to methionine (H3K27M) are seen in over 2/3 of DMGs, and are associated with a worse prognosis. Due to the anatomical location of DMG, traditional biopsy carries risk for neurologic injury as it requires penetration of vital midline structures. Further, radiographic (MRI) monitoring of DMG often shows nonspecific changes, which makes therapeutic monitoring difficult. This indicates a critical need for more minimally invasive methods, such as liquid biopsy, to understand, diagnose, and monitor H3K27M DMG. Here, we review the use of all modalities to date to detect biomarkers of H3K27M in cerebrospinal fluid (CSF), blood, and urine, and compare their effectiveness in detection, diagnosis, and monitoring treatment response. We provide specific detail of recent efforts to monitor CSF and plasma H3K27M cell-free DNA in patients undergoing therapy with the imipridone ONC201. Lastly, we discuss the future of therapeutic monitoring of H3K27M-DMG, including biomarkers such as mitochondrial DNA, mutant and modified histones, and novel sequencing-based approaches for improved detection methods.
Collapse
Affiliation(s)
- Jina Patel
- Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Rayan Aittaleb
- Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Robert Doherty
- Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Ananya Gera
- Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Benison Lau
- Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Dana Messinger
- Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Jack Wadden
- Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Andrea Franson
- Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan, USA
| | | | - Carl Koschmann
- Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan, USA
| |
Collapse
|
48
|
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
|
49
|
Xin DE, Liao Y, Rao R, Ogurek S, Sengupta S, Xin M, Bayat AE, Seibel WL, Graham RT, Koschmann C, Lu QR. Chaetocin-mediated SUV39H1 inhibition targets stemness and oncogenic networks of diffuse midline gliomas and synergizes with ONC201. Neuro Oncol 2024; 26:735-748. [PMID: 38011799 PMCID: PMC10995509 DOI: 10.1093/neuonc/noad222] [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: 05/22/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Diffuse intrinsic pontine gliomas (DIPG/DMG) are devastating pediatric brain tumors with extraordinarily limited treatment options and uniformly fatal prognosis. Histone H3K27M mutation is a common recurrent alteration in DIPG and disrupts epigenetic regulation. We hypothesize that genome-wide H3K27M-induced epigenetic dysregulation makes tumors vulnerable to epigenetic targeting. METHODS We performed a screen of compounds targeting epigenetic enzymes to identify potential inhibitors for the growth of patient-derived DIPG cells. We further carried out transcriptomic and genomic landscape profiling including RNA-seq and CUT&RUN-seq as well as shRNA-mediated knockdown to assess the effects of chaetocin and SUV39H1, a target of chaetocin, on DIPG growth. RESULTS High-throughput small-molecule screening identified an epigenetic compound chaetocin as a potent blocker of DIPG cell growth. Chaetocin treatment selectively decreased proliferation and increased apoptosis of DIPG cells and significantly extended survival in DIPG xenograft models, while restoring H3K27me3 levels. Moreover, the loss of H3K9 methyltransferase SUV39H1 inhibited DIPG cell growth. Transcriptomic and epigenomic profiling indicated that SUV39H1 loss or inhibition led to the downregulation of stemness and oncogenic networks including growth factor receptor signaling and stemness-related programs; however, D2 dopamine receptor (DRD2) signaling adaptively underwent compensatory upregulation conferring resistance. Consistently, a combination of chaetocin treatment with a DRD2 antagonist ONC201 synergistically increased the antitumor efficacy. CONCLUSIONS Our studies reveal a therapeutic vulnerability of DIPG cells through targeting the SUV39H1-H3K9me3 pathway and compensatory signaling loops for treating this devastating disease. Combining SUV39H1-targeting chaetocin with other agents such as ONC201 may offer a new strategy for effective DIPG treatment.
Collapse
Affiliation(s)
- Dazhuan Eric Xin
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Yunfei Liao
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Rohit Rao
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Sean Ogurek
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Soma Sengupta
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Mei Xin
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Arman Esshaghi Bayat
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - William L Seibel
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Richard T Graham
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Carl Koschmann
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Q Richard Lu
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
50
|
Papangelopoulou D, Bison B, Behrens L, Bailey S, Ansari M, Ehlert K, Martinez OC, Kramm CM, Morales La Madrid A, von Bueren AO. Brain stem tumors in children less than 3 months: Clinical and radiologic findings of a rare disease. Childs Nerv Syst 2024; 40:1053-1064. [PMID: 38376530 DOI: 10.1007/s00381-023-06272-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/26/2023] [Indexed: 02/21/2024]
Abstract
PURPOSE Brain stem tumors in children < 3 months at diagnosis are extremely rare. Our aim is to study a retrospective cohort to improve the understanding of the disease course and guide patient management. METHODS This is a multicenter retrospective analysis across the European Society for Pediatric Oncology SIOP-E HGG/DIPG Working Group linked centers, including patients with a brainstem tumor diagnosed between 2009 and 2020 and aged < 3 months at diagnosis. Clinical data were collected, and imaging characteristics were analyzed blindly and independently by two neuroradiologists. RESULTS Five cases were identified. No patient received any therapy. The epicenter of two tumors was in the medulla oblongata alone and in the medulla oblongata and the pons in three. For patients with tumor in equal parts in the medulla oblongata and the pons (n = 3), the extension at diagnosis involved the spinal cord; for the two patients with the tumor epicenter in the medulla oblongata alone (n = 2), the extension at diagnosis included the pons (n = 2) and the spinal cord (n = 1). Biopsy was performed in one patient identifying a pilocytic astrocytoma. Two patients died. In one patient, autopsy revealed a high-grade glioma (case 3). Three survivors showed either spontaneous tumor regression (n = 2) or stable disease (n = 1). Survivors were followed up for 10, 7, and 0.6 years, respectively. One case had the typical imaging characteristics of a dorsal exophytic low-grade glioma. CONCLUSIONS No patient fulfilled the radiologic criteria defining a high-grade glioma. Central neuroradiological review and biopsy may provide useful information regarding the patient management.
Collapse
Affiliation(s)
- Danai Papangelopoulou
- Department of Pediatrics, Gynecology and Obstetrics, Division of General Pediatrics, Pediatric Hematology and Oncology Unit, University Hospitals of Geneva, Geneva, Switzerland
- Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland
| | - Brigitte Bison
- Diagnostic and Interventional Neuroradiology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Lars Behrens
- Diagnostic and Interventional Neuroradiology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Simon Bailey
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Marc Ansari
- Department of Pediatrics, Gynecology and Obstetrics, Division of General Pediatrics, Pediatric Hematology and Oncology Unit, University Hospitals of Geneva, Geneva, Switzerland
- Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland
| | - Karoline Ehlert
- Department of Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| | | | - Christof M Kramm
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Goettingen, Goettingen, Germany
| | | | - Andre O von Bueren
- Department of Pediatrics, Gynecology and Obstetrics, Division of General Pediatrics, Pediatric Hematology and Oncology Unit, University Hospitals of Geneva, Geneva, Switzerland.
- Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland.
| |
Collapse
|