1
|
Faison SL, Batonga J, Arumugham T, Bartkus A, Morrison M, Mullin MJ, Tippin T, Naderer O. A Phase 1 Randomized Study to Evaluate the Safety, Tolerability, and Pharmacokinetics of Single Escalating Oral Doses of Dordaviprone and the Effects of Food on the Bioavailability of Dordaviprone in Healthy Adult Subjects. Clin Pharmacol Drug Dev 2025; 14:382-390. [PMID: 39817482 DOI: 10.1002/cpdd.1512] [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: 11/13/2024] [Accepted: 12/29/2024] [Indexed: 01/18/2025]
Abstract
Dordaviprone (ONC201) is a novel, small molecule imipridone with antitumor effects in glioma patients. This study evaluated the pharmacokinetics and safety of dordaviprone following single escalating doses (Part A), as a capsule content mixed with applesauce or Gatorade (sports drink) [Part B1]), and with or without food [Part B2]. The most common treatment-emergent adverse events pooled across study parts (Parts A, B1, and B2) were headache, dizziness, and headache, respectively; all were mild. Systemic dordaviprone exposure increased dose proportionally following administration of 125-625 mg of dordaviprone. Following dordaviprone capsule contents sprinkled on applesauce or dissolved in sports drink, the geometric mean ratios, and 90% confidence intervals (CIs) of the dordaviprone area under the concentration versus time curve (AUC) fell within the bioequivalence (BE) limits of 80.00%-125.00%; however, for Cmax the 90% CI lower limit (0.70) fell below BE limits when sprinkled on applesauce. The geometric mean ratios and 90% CIs of dordaviprone administered under fed versus fasted conditions fell within BE limits of 80.00%-125.00% for the AUC, indicating no food effect on total exposure; however, maximum concentration (Cmax) (90% CI 0.55, 0.67) fell below BE limits.
Collapse
Affiliation(s)
| | | | | | - Angela Bartkus
- Chimerix, Inc., Clinical Pharmacology and Translational Medicine, Durham, NC, USA
| | - Marion Morrison
- Chimerix, Inc., Clinical Pharmacology and Translational Medicine, Durham, NC, USA
| | - Mark J Mullin
- Chimerix, Inc., Clinical Pharmacology and Translational Medicine, Durham, NC, USA
| | - Tim Tippin
- Chimerix, Inc., Clinical Pharmacology and Translational Medicine, Durham, NC, USA
| | - Odin Naderer
- Chimerix, Inc., Clinical Pharmacology and Translational Medicine, Durham, NC, USA
| |
Collapse
|
2
|
Sun M, Zheng J, Gong L, Li Z, Wang L. Personalized combination therapy for diffuse midline glioma: A case report. Oncol Lett 2025; 29:234. [PMID: 40151419 PMCID: PMC11948954 DOI: 10.3892/ol.2025.14980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/06/2025] [Indexed: 03/29/2025] Open
Abstract
The present study aimed to analyze the efficacy of personalized combination therapy for patients with H3K27M mutant diffuse midline glioma (DMG) so as to explore new treatment options for further clinical research. The clinical data and prognosis of a patient with H3K27M mutant DMG are summarized and discussed in the context of the relevant literature. The patient was a 20-year-old female diagnosed with DMG treated with a combination of surgery, radiotherapy, chemotherapy, electric field therapy, immunotherapy and targeted therapy. An overall survival time of 28 months was achieved. In summary, personalized treatment strategies are expected to provide longer-lasting survival benefits for patients with DMG.
Collapse
Affiliation(s)
- Mengqi Sun
- Department of Radiation Oncology, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, Guangdong 518000, P.R. China
| | - Jizhou Zheng
- Department of Radiation Oncology, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, Guangdong 518000, P.R. China
| | - Long Gong
- Department of Radiation Oncology, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, Guangdong 518000, P.R. China
| | - Zihuang Li
- Department of Radiation Oncology, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, Guangdong 518000, P.R. China
| | - Lijun Wang
- Department of Radiation Oncology, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, Guangdong 518000, P.R. China
| |
Collapse
|
3
|
Stockwell CA, Thang M, Kram DE, Satterlee AB, Hingtgen S. Therapeutic approaches for targeting the pediatric brain tumor microenvironment. Drug Deliv Transl Res 2025:10.1007/s13346-025-01839-3. [PMID: 40257744 DOI: 10.1007/s13346-025-01839-3] [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] [Accepted: 03/14/2025] [Indexed: 04/22/2025]
Abstract
Central nervous system (CNS) tumors are the most frequent solid malignant tumors in pediatric patients and are the leading cause of tumor-related death in children. Treatment for this heterogeneous group of tumors consists of various combinations of safe maximal surgical resection, chemotherapy, and radiation therapy which offer a cure for some children but often cause debilitating adverse late effects in others. While therapies targeting the tumor microenvironment (TME) like immune checkpoint inhibition (ICI) have been successful in treating some cancers, these therapies failed to exhibit treatment efficacy in the majority of pediatric brain tumors in the clinic. Importantly, the pediatric TME is unique and distinct from adult brain tumors and designing therapies to effectively target these tumors requires understanding the unique biology of pediatric brain tumors and the use of translational models that recapitulate the TME. Here we describe the TME of medulloblastoma (MB) and diffuse midline glioma (DMG), specifically diffuse intrinsic pontine glioma (DIPG), and further present the current drug delivery approaches and clinical administration routes targeting the TME in these tumors, including preclinical and clinical studies.
Collapse
Affiliation(s)
- Caroline A Stockwell
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Morrent Thang
- Neuroscience Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David E Kram
- Division of Pediatric Hematology-Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew B Satterlee
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Eshelman Innovation, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shawn Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
4
|
Teffera Y, Tippin T, Bartkus A, de Castro FA, Faison SL, Morrison M, Mullin MJ, Naderer O. A Phase 1 Study to Evaluate the Absorption, Metabolism, and Disposition of Dordaviprone in Healthy Adult Participants. Clin Pharmacol Drug Dev 2025. [PMID: 40243222 DOI: 10.1002/cpdd.1541] [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: 01/31/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
Dordaviprone (ONC201) is a novel, small-molecule imipridone with antitumor activity in patients with a glioma. Six healthy male participants received a single 625-mg (100-µCi) oral dose of [14C]-dordaviprone. Blood, plasma, urine, and feces were collected up to 288 hours after dosing and analyzed by liquid scintillation counting. Metabolite profiles were evaluated using liquid chromatography-radiometric detection, and metabolite identification was accomplished by liquid chromatography with tandem mass spectrometry. Concentrations of drug-derived radioactivity in blood and plasma peaked at 1 hour after dosing and were below the limit of quantitation by 72 hours (whole blood) to 96 hours (plasma) after dosing. Seventy-one percent of the administered radioactivity was recovered in urine and 20% in feces. In plasma, the major circulating compounds were dordaviprone and the inactive metabolite ONC207, each contributing approximately one third of the total radioactivity area under the curve. Of the 19 metabolites identified in plasma, no other single metabolite contributed more than 10% to the total radioactivity area under the curve. Unchanged dordaviprone was a minor component in excreta (less than 0.3%), with multiple metabolites identified in urine and feces. Given the lack of dordaviprone in excreta and the metabolites formed, the primary route for dordaviprone elimination was through urinary excretion of oxidative metabolites.
Collapse
|
5
|
Liu C, Liu Y, Lin H, Zhang C, Zhang B, Song H, Fan X, Lyu Y, Yang H, Mao Y. Multi-omics landscape of alternative splicing in diffuse midline glioma reveals immune- and neural-driven subtypes with implications for spliceosome-targeted therapy. Front Immunol 2025; 16:1587009. [PMID: 40308585 PMCID: PMC12040961 DOI: 10.3389/fimmu.2025.1587009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction H3K27-altered diffuse midline glioma (DMG) is a highly aggressive glioma subtype, accounting for approximately 60% of pediatric high-grade gliomas, with a median survival of less than 12 months. Due to its predominant localization in the brainstem, conventional surgical resection is often unfeasible, underscoring the urgent need for alternative therapeutic strategies. While previous studies on DMG have primarily focused on regulatory mechanisms at the protein level, the role of alternative splicing in DMG remains largely unexplored. Given its potential impact on gene regulation and tumor progression, a comprehensive analysis of alternative splicing could provide novel insights into targeted or immune therapeutic strategies, complementing existing transcriptomic studies of DMG. Methods To investigate the alternative splicing landscape of DMG, we performed transcriptome sequencing (RNA-seq) on patient-derived H3WT and H3K27-altered DMG cell lines, integrating these data with RNA-seq and single-cell transcriptomic (scRNA-seq) datasets from published sources. This comprehensive approach enabled us to delineate the alternative splicing landscape of H3K27-altered DMG and validate its distinct features at the cellular level. Results Our multi-omics analysis revealed significant transcriptional alterations in H3K27-altered DMG compared to H3WT DMG, particularly in pathways related to neuro-regulation, metabolism, and immunity. Further in-depth analysis identified extensive alternative splicing changes in H3K27-altered DMG, predominantly associated with RNA modifications and key alterations in extracellular matrix and nucleotide metabolism. Integrating these findings, we characterized five RNA-associated proteins that enabled a binary classification of DMG into neural and immune subtypes, with each subtype exhibiting distinct prognostic and transcriptomic features. Notably, we identified RALYL as a potential key regulator in DMG progression. Discussion Our findings indicate that H3K27-altered DMG exhibits significant alternative splicing alterations, which play crucial roles in tumorigenesis and progression. Additionally, our study identified an RNA-binding protein-based classification of DMG and characterized RALYL as a potential regulatory factor, highlighting its potential as a novel therapeutic target.
Collapse
Affiliation(s)
- Chaxian Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yue Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hao Lin
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chufan Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bilong Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Haikun Song
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaomin Fan
- Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Lyu
- Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Mayr L, Neyazi S, Schwark K, Trissal M, Beck A, Labelle J, Eder SK, Weiler-Wichtl L, Marques JG, de Biagi-Junior CAO, Lo Cascio C, Chapman O, Sridhar S, Kenkre R, Dutta A, Wang S, Wang J, Hack O, Nascimento A, Nguyen CM, Castellani S, Rozowsky JS, Groves A, Panditharatna E, Cruzeiro GAV, Haase RD, Tabatabai K, Madlener S, Wadden J, Adam T, Kong S, Miclea M, Patel T, Bruckner K, Senfter D, Lämmerer A, Supko J, Guntner AS, Palova H, Neradil J, Stepien N, Lötsch-Gojo D, Berger W, Leiss U, Rosenmayr V, Dorfer C, Dieckmann K, Peyrl A, Azizi AA, Baumgartner A, Slaby O, Pokorna P, Clark LM, Cameron A, Nguyen QD, Wakimoto H, Dubois F, Greenwald NF, Bandopadhayay P, Beroukhim R, Ligon K, Kramm C, Bronsema A, Bailey S, Stucklin AG, Mueller S, Skrypek M, Martinez N, Bowers DC, Jones DTW, Jones C, Jäger N, Sterba J, Müllauer L, Haberler C, Kumar-Sinha C, Chinnaiyan A, Mody R, Chavez L, Furtner J, Koschmann C, Gojo J, Filbin MG. Effective targeting of PDGFRA-altered high-grade glioma with avapritinib. Cancer Cell 2025; 43:740-756.e8. [PMID: 40086436 DOI: 10.1016/j.ccell.2025.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/27/2024] [Accepted: 02/12/2025] [Indexed: 03/16/2025]
Abstract
PDGFRA is crucial to tumorigenesis and frequently genomically altered in high-grade glioma (HGG). In a comprehensive dataset of pediatric HGG (n = 261), we detect PDGFRA mutations and/or amplifications in 15% of cases, suggesting PDGFRA as a therapeutic target. We reveal that the PDGFRA/KIT inhibitor avapritinib shows (1) selectivity for PDGFRA inhibition, (2) distinct patterns of subcellular effects, (3) in vitro and in vivo activity in patient-derived HGG models, and (4) effective blood-brain barrier penetration in mice and humans. Furthermore, we report preliminary clinical real-world experience using avapritinib in pediatric and young adult patients with predominantly recurrent/refractory PDGFRA-altered HGG (n = 8). Our early data demonstrate that avapritinib is well tolerated and results in radiographic response in 3/7 cases, suggesting a potential role for avapritinib in the treatment of HGG with specific PDGFRA alterations. Overall, these translational results underscore the therapeutic potential of PDGFRA inhibition with avapritinib in HGG.
Collapse
Affiliation(s)
- Lisa Mayr
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA; Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Sina Neyazi
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Kallen Schwark
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maria Trissal
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Alexander Beck
- Center for Neuropathology and Prion Research, Ludwig Maximilians University Munich, Faculty of Medicine, Muenchen, 80539 Bayern, Germany
| | - Jenna Labelle
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Sebastian K Eder
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA; St. Anna Children's Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna and St. Anna Children's Cancer Research Institute (CCRI), 1090 Vienna, Austria
| | - Liesa Weiler-Wichtl
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Joana G Marques
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Carlos A O de Biagi-Junior
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Costanza Lo Cascio
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Owen Chapman
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA 92037, USA
| | - Sunita Sridhar
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA 92037, USA
| | - Rishaan Kenkre
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA 92037, USA
| | - Aditi Dutta
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA 92037, USA
| | - Shanqing Wang
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA 92037, USA
| | - Jessica Wang
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA 92037, USA
| | - Olivia Hack
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Andrezza Nascimento
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Cuong M Nguyen
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Sophia Castellani
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Jacob S Rozowsky
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Andrew Groves
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Eshini Panditharatna
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Gustavo Alencastro Veiga Cruzeiro
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Rebecca D Haase
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Kuscha Tabatabai
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Sibylle Madlener
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Jack Wadden
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Tiffany Adam
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Seongbae Kong
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Madeline Miclea
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Tirth Patel
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Katharina Bruckner
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; Department of Neurosurgery, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Daniel Senfter
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Anna Lämmerer
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; Research Cluster "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, 1090 Vienna, Austria
| | - Jeffrey Supko
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Armin S Guntner
- Institute for Analytical and General Chemistry, Johannes Kepler University, 4040 Linz, Austria
| | - Hana Palova
- Central European Institute of Technology, Masaryk University, 60177 Brno, Czech Republic
| | - Jakub Neradil
- Department of Experimental Biology, Faculty of Science, Masaryk University, 61137 Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Natalia Stepien
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Daniela Lötsch-Gojo
- Department of Neurosurgery, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Walter Berger
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; Research Cluster "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, 1090 Vienna, Austria
| | - Ulrike Leiss
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Verena Rosenmayr
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Christian Dorfer
- Department of Neurosurgery, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Karin Dieckmann
- Department of Radiotherapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Andreas Peyrl
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Amedeo A Azizi
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Alicia Baumgartner
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA; Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, 60177 Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Petra Pokorna
- Central European Institute of Technology, Masaryk University, 60177 Brno, Czech Republic
| | - Louise M Clark
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute, Boston, MA 02210, USA
| | - Amy Cameron
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute, Boston, MA 02210, USA
| | - Quang-De Nguyen
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute, Boston, MA 02210, USA
| | - Hiroaki Wakimoto
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Frank Dubois
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Noah F Greenwald
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Cancer Biology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02115, USA
| | - Pratiti Bandopadhayay
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Rameen Beroukhim
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Cancer Biology and Medical Oncology, Dana-Farber Cancer Institute, and Harvard Medical School, Boston, MA 02115, USA
| | - Keith Ligon
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Christof Kramm
- Division of Pediatric Hematology and Oncology, University Medical Center Göttingen, 37099 Göttingen, Germany
| | - Annika Bronsema
- Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Simon Bailey
- Great North Childrens Hospital and Newcastle University, Newcastle upon Tyne, UK; Newcastle Hospitals NHS Foundation Trust, NE1 4LP Newcastle, UK
| | - Ana Guerreiro Stucklin
- Department of Oncology and Children's Research Center, University Children's Hospital Zurich, 8008 Zurich, Switzerland
| | - Sabine Mueller
- Departments of Pediatrics, Neurology, and Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Mary Skrypek
- Department of Pediatric Hematology-Oncology, Children's Minnesota, Minneapolis, MN 55404, USA
| | - Nina Martinez
- Department of Neurology & Neurological Surgery, Jefferson University, Philadelphia, PA 19107, USA
| | - Daniel C Bowers
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - David T W Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ), 69120 Heidelberg, Germany; Pediatric Glioma Research Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Chris Jones
- Division of Molecular Pathology, Institute of Cancer Research, SM2 5NG London, UK
| | - Natalie Jäger
- Hopp Children's Cancer Center Heidelberg (KiTZ) & Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Jaroslav Sterba
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 662630 Brno, Czech Republic
| | - Leonhard Müllauer
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria
| | - Christine Haberler
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Chandan Kumar-Sinha
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Arul Chinnaiyan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Rajen Mody
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Lukas Chavez
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA 92037, USA
| | - Julia Furtner
- Division of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria; Research Center of Medical Image Analysis and Artificial Intelligence, Danube Private University, 3500 Krems an der Donau, Austria
| | - Carl Koschmann
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Johannes Gojo
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria.
| | - Mariella G Filbin
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
7
|
Shen ZP, Zhang ZY, Li N, Xu L, Chen Y. Targeted therapy for pediatric glioma: RAF(t)ing in the molecular era. World J Pediatr 2025:10.1007/s12519-025-00889-4. [PMID: 40227462 DOI: 10.1007/s12519-025-00889-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Pediatric gliomas are the most frequently occurring central nervous system tumors in children. While targeted therapies have been widely applied in the treatment of many adult cancers, their use in pediatric gliomas has lagged behind. However, recent advances in multiomics profiling of pediatric gliomas, coupled with the approval of inhibitors against Raf serine/threonine kinase (RAF), isocitrate dehydrogenase 1/2 (IDH1/2) and neurotrophic receptor tyrosine kinase (NTRK), have spurred significant progress in this field. In light of these developments, this review aims to provide a comprehensive overview of current advancements and the evolving landscape of targeted therapeutic strategies and approaches for pediatric gliomas. DATA SOURCES Data analyzed in this study were obtained from the literature from PubMed, as well as other online databases and websites, including ClinicalTrials.gov and the Pediatric Neuro-Oncology Consortium. RESULTS Based on findings from multiomics profiling, significant insights have been gained into the genetic and molecular landscape of pediatric gliomas, enabling the identification of key mutations and potentially targetable lesions. These advancements provide rationales for the development of more precise treatment strategies and targeted therapies. Recent approvals of targeted therapies and ongoing clinical trials in pediatric gliomas are converging on the targeting of key signaling molecules and metabolic pathways. CONCLUSIONS In the molecular era, targeted therapies offer new hope for more effective and personalized treatment options for pediatric glioma patients. By developing and tailoring treatments to target specific molecular and metabolic vulnerabilities, targeted therapies have the potential to improve the clinical management of pediatric gliomas, ultimately enhancing both the treatment experience and overall prognosis of these patients.
Collapse
Affiliation(s)
- Zhi-Peng Shen
- Department of Neurosurgery, Children's Hospital Zhejiang University School of Medicine, Hangzhou, 310052, China
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Zhong-Yuan Zhang
- Department of Neurosurgery, Children's Hospital Zhejiang University School of Medicine, Hangzhou, 310052, China
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Nan Li
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Liang Xu
- Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China.
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
- Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| | - Ye Chen
- Department of Neurosurgery, Children's Hospital Zhejiang University School of Medicine, Hangzhou, 310052, China.
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou, 310052, China.
- Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
8
|
Dasgupta P, Puduvalli VK. Diversity of metabolic features and relevance to clinical subtypes of gliomas. Semin Cancer Biol 2025; 112:126-134. [PMID: 40194749 DOI: 10.1016/j.semcancer.2025.03.008] [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: 08/14/2024] [Revised: 02/13/2025] [Accepted: 03/31/2025] [Indexed: 04/09/2025]
Abstract
Gliomas carry a dismal prognosis and have proven difficult to treat. Current treatments and efforts to target individual signaling pathways have failed. This is thought to be due to genetic and epigenetic heterogeneity and resistance. Therefore, interest has grown in developing a deeper understanding of the metabolic alterations that represent drivers and dependencies in gliomas. Therapies that target glioma-specific metabolic dependencies overcome the challenges of disease heterogeneity. Here, we present the diverse metabolic features of each current clinical subtype of glioma. We believe that this approach will enable the development of novel strategies to specifically target the various clinical and molecular subtypes of glioma using these metabolic features.
Collapse
Affiliation(s)
- Pushan Dasgupta
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA.
| | - Vinay K Puduvalli
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA.
| |
Collapse
|
9
|
Faison SL, Batonga J, Arumugham T, Bartkus A, Morrison M, Mullin MJ, Tippin T, Naderer O. Relative Bioavailability of Dordaviprone (ONC201) is Not Affected by Co-Administration of the Proton-Pump Inhibitor Rabeprazole. J Clin Pharmacol 2025; 65:520-526. [PMID: 39577877 DOI: 10.1002/jcph.6163] [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: 08/09/2024] [Accepted: 10/30/2024] [Indexed: 11/24/2024]
Abstract
Dordaviprone (ONC201) is a novel, orally administered, anti-cancer, small molecule imipridone with demonstrated antitumor effects in patients with glioma. Dordaviprone in vitro solubility is significantly reduced at pH >4.5. Concomitant use of acid reducing agents (ARAs) may therefore impact dordaviprone solubility and bioavailability. This open-label, single-sequence, three-period crossover study evaluated the effect of proton-pump inhibitor rabeprazole on dordaviprone pharmacokinetics (PK). Periods were consecutive and comprised of period 1 (days 1-3), period 2 (days 4-9), and period 3 (days 10-13). In period 1, participants received a single oral 625 mg dose of dordaviprone on day 1. In period 2, participants received six consecutive days of QD 20 mg rabeprazole alone. In period 3, patients received one oral dose of 20 mg rabeprazole (the seventh consecutive daily dose), followed 2 h later by a single 625 mg dordaviprone oral dose. PK blood samples were collected and analyzed from pre-dose 72 h following dordaviprone administration in periods 1 and 3. Dordaviprone exposure PK parameters were similar following administration of dordaviprone alone or with rabeprazole. Geometric mean ratios and 90% CIs for dordaviprone exposure parameters with and without rabeprazole following dordaviprone administration fell within bioequivalence limits of 80.00%-125.00% for Cmax (97.19% [86.43-109.28]), AUClast (102.21% [95.19-109.75]), and AUCinf (102.27% [95.21-109.86]), indicating no effect of multiple oral doses of rabeprazole on dordaviprone relative bioavailability. Six of the 16 participants reported treatment-emergent adverse events (TEAEs); dordaviprone-related TEAEs were reported by three participants and were limited to mild nausea and dizziness. No dordaviprone dose adjustment or ARA treatment modification is warranted.
Collapse
|
10
|
Zhong S, Zuo J, Fu X, Wu C, Liu R, Huang Z, Li S. Leptomeningeal dissemination in H3 K27M- mutant diffuse midline gliomas: clinical characteristics, risk factors, and prognostic insights. J Neurooncol 2025; 172:437-445. [PMID: 39812935 PMCID: PMC11937158 DOI: 10.1007/s11060-024-04933-7] [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: 11/23/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025]
Abstract
PURPOSE This study aimed to describe the incidence, clinical and pathological features, and outcomes of H3 K27M- mutant Diffuse Midline Glioma (DMG) patients with leptomeningeal dissemination (LMD) and systematically investigate the predictive and prognostic factors to clarify the response to treatment after the onset of LMD. METHODS A total of 304 patients diagnosed with DMG from October 17, 2017, to October 17, 2023, were enrolled in this study, of which 32 patients were diagnosed with LMD. Logistic regression analyses were conducted to identify the predictors of LMD, including clinical, molecular, and imaging data. Univariable and multivariable cox regression analyses were used for overall survival (OS) and post-LMD survival (PLS) analysis. RESULTS The median OS and PLS were 12.5 and 8.0 months respectively. Tumor with contrast-enhanced lesions reaching ependyma (Ventricular contact type I) was the only independent risk factor for LMD. Male sex and ventricular contact type I were independent risk factors for primary LMD. In all LMD patients, Karnofsky Performance Status (KPS) of ≥ 90 and radiotherapy were statistically significantly associated with longer OS, and primary LMD was significantly associated with shorter OS. Supratentorial location and chemotherapy after LMD diagnosis were independent favorable prognostic factors on PLS. In primary LMD subgroup analysis, radiotherapy was the only independent favorable prognostic factor on OS. CONCLUSIONS The association between contrast-enhanced lesions and ventricular involvement is an independent predictive factor for LMD in DMG patients. Radiotherapy and preoperative KPS may contribute to improved overall survival in these patients. Chemotherapy is a potential treatment option following an LMD diagnosis.
Collapse
Affiliation(s)
- Shuai Zhong
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Jinyi Zuo
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Xiaojun Fu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Chenxing Wu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Rui Liu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Zheng Huang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Shouwei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China.
| |
Collapse
|
11
|
McVeigh L, Patel T, Miclea M, Schwark K, Ajaero D, Momen F, Clausen M, Adam T, Aittaleb R, Wadden J, Lau B, Franson AT, Koschmann C, Marupudi NI. Updates in Diagnostic Techniques and Experimental Therapies for Diffuse Intrinsic Pontine Glioma. Cancers (Basel) 2025; 17:931. [PMID: 40149267 PMCID: PMC11940218 DOI: 10.3390/cancers17060931] [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: 01/25/2025] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a rare but extremely malignant central nervous system tumor primarily affecting children that is almost universally fatal with a devastating prognosis of 8-to-12-month median survival time following diagnosis. Traditionally, DIPG has been diagnosed via MR imaging alone and treated with palliative radiation therapy. While performing surgical biopsies for these patients has been controversial, in recent years, advancements have been made in the safety and efficacy of surgical biopsy techniques, utilizing stereotactic, robotics, and intraoperative cranial nerve monitoring as well as the development of liquid biopsies that identify tumor markers in either cerebrospinal fluid or serum. With more molecular data being collected from these tumors due to more frequent biopsies being performed, multiple treatment modalities including chemotherapy, radiation therapy, immunotherapy, and epigenetic modifying agents continue to be developed. Numerous recent clinical trials have been completed or are currently ongoing that have shown promise in extending survival for patients with DIPG. Focused ultrasound (FUS) has also emerged as an additional promising adjunct invention used to increase the effectiveness of therapeutic agents. In this review, we discuss the current evidence to date for these advancements in the diagnosis and treatment of DIPG.
Collapse
Affiliation(s)
- Luke McVeigh
- Department of Neurosurgery, Michigan Medicine, Ann Arbor, MI 48109, USA;
| | - Tirth Patel
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Madeline Miclea
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Kallen Schwark
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Diala Ajaero
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Fareen Momen
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Madison Clausen
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Tiffany Adam
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Rayan Aittaleb
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Jack Wadden
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Benison Lau
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Andrea T. Franson
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Carl Koschmann
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Neena I. Marupudi
- Department of Neurosurgery, Michigan Medicine, Ann Arbor, MI 48109, USA;
| |
Collapse
|
12
|
Kampers LFC, Metselaar DS, Vinci M, Scirocchi F, Veldhuijzen van Zanten S, Eyrich M, Biassoni V, Hulleman E, Karremann M, Stücker W, Van Gool SW. The Complexity of Malignant Glioma Treatment. Cancers (Basel) 2025; 17:879. [PMID: 40075726 PMCID: PMC11899524 DOI: 10.3390/cancers17050879] [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: 01/22/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Malignant glioma is a highly aggressive, therapeutically non-responsive, and deadly disease with a unique tumor microenvironment (TME). Of the 14 currently recognized and described cancer hallmarks, five are especially implicated in malignant glioma and targetable with repurposed drugs: cancer stem-like cells, in general, and glioma stem-like cells in particular (GSCs), vascularization and hypoxia, metabolic reprogramming, tumor-promoting inflammation and sustained proliferative signaling. Each hallmark drives malignant glioma development, both individually and through interactions with other hallmarks, in which the TME plays a critical role. To combat the aggressive malignant glioma spatio-temporal heterogeneity driven by TME interactions, and to overcome its therapeutic challenges, a combined treatment strategy including anticancer therapies, repurposed drugs and multimodal immunotherapy should be the aim for future treatment approaches.
Collapse
Affiliation(s)
| | - Dennis S. Metselaar
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (D.S.M.); (S.V.v.Z.); (E.H.)
- Hopp Children’s Cancer Center Heidelberg (KiTZ), 69120 Heidelberg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Consortium (DKTK), 69120 Heidelberg, Germany
| | - Maria Vinci
- Bambino Gesu’ Children’s Hospital-IRCCS, 00165 Rome, RM, Italy; (M.V.); (F.S.)
| | - Fabio Scirocchi
- Bambino Gesu’ Children’s Hospital-IRCCS, 00165 Rome, RM, Italy; (M.V.); (F.S.)
| | - Sophie Veldhuijzen van Zanten
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (D.S.M.); (S.V.v.Z.); (E.H.)
- Department of Radiology & Nuclear Medicine, Erasmus MC—University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands
- Brain Tumour Centre, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | | | - Veronica Biassoni
- Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milano, MI, Italy;
| | - Esther Hulleman
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (D.S.M.); (S.V.v.Z.); (E.H.)
| | | | - Wilfried Stücker
- Immun-Onkologisches Zentrum Köln, 50674 Köln, Germany; (L.F.C.K.); (W.S.)
| | | |
Collapse
|
13
|
Venneti S, Kawakibi AR, Ji S, Waszak SM, Sweha SR, Mota M, Pun M, Deogharkar A, Chung C, Tarapore RS, Ramage S, Chi A, Wen PY, Arrillaga-Romany I, Batchelor TT, Butowski NA, Sumrall A, Shonka N, Harrison RA, de Groot J, Mehta M, Hall MD, Daghistani D, Cloughesy TF, Ellingson BM, Beccaria K, Varlet P, Kim MM, Umemura Y, Garton H, Franson A, Schwartz J, Jain R, Kachman M, Baum H, Burant CF, Mottl SL, Cartaxo RT, John V, Messinger D, Qin T, Peterson E, Sajjakulnukit P, Ravi K, Waugh A, Walling D, Ding Y, Xia Z, Schwendeman A, Hawes D, Yang F, Judkins AR, Wahl D, Lyssiotis CA, Nava DDL, Alonso MM, Eze A, Spitzer J, Schmidt SV, Duchatel RJ, Dun MD, Cain JE, Jiang L, Stopka SA, Baquer G, Regan MS, Filbin MG, Agar NYR, Zhao L, Kumar-Sinha C, Mody R, Chinnaiyan A, Kurokawa R, Pratt D, Yadav VN, Grill J, Kline C, Mueller S, Resnick A, Nazarian J, Allen JE, Odia Y, Gardner SL, Koschmann C. Correction: Clinical Efficacy of ONC201 in H3K27M-Mutant Diffuse Midline Gliomas Is Driven by Disruption of Integrated Metabolic and Epigenetic Pathways. Cancer Discov 2025; 15:657. [PMID: 40025951 PMCID: PMC11873718 DOI: 10.1158/2159-8290.cd-25-0208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 01/30/2025] [Indexed: 03/04/2025]
|
14
|
Nuño MM, Pugh SL, Ji L, Piao J, Dignam JJ, Steingrimsson JA. On the use of external controls in clinical trials. J Natl Cancer Inst Monogr 2025; 2025:30-34. [PMID: 39989040 PMCID: PMC11848027 DOI: 10.1093/jncimonographs/lgae046] [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: 08/09/2024] [Revised: 09/09/2024] [Accepted: 10/01/2024] [Indexed: 02/25/2025] Open
Abstract
Externally controlled trials have commonly been used when conducting a randomized controlled trial (RCT) is not feasible or ethical. By allowing the study of new treatments, use of external controls can lead to accelerated advances in the management of rare diseases. The use of external controls, however, introduces new challenges due to potential differences between the population the external controls are enrolled from and the population the patients on the new trial are enrolled from. Some differences include, but are not limited to, differences in how patients are diagnosed and treated, differences in the case mix of the underlying populations, differences in the ability to measure outcomes, and differences in data collection. We discuss the potential benefits and challenges of externally controlled trials, as well as strategies to mitigate bias, including the estimand and target-trial emulation framework. We also provide a brief overview of statistical methodology commonly used in these settings. We note that although the strategies presented may help mitigate some of these challenges, they cannot replace an RCT framework, and investigators should be aware of the potential limitations of externally controlled trials.
Collapse
Affiliation(s)
- Michelle M Nuño
- Children’s Oncology Group, Monrovia, CA 91016, United States
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA 90032, United States
| | - Stephanie L Pugh
- NRG Oncology Statistics and Data Management Center, American College of Radiology, Philadelphia, PA 19103, United States
| | - Lingyun Ji
- Children’s Oncology Group, Monrovia, CA 91016, United States
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA 90032, United States
| | - Jin Piao
- Children’s Oncology Group, Monrovia, CA 91016, United States
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA 90032, United States
| | - James J Dignam
- NRG Oncology Statistics and Data Management Center, American College of Radiology, Philadelphia, PA 19103, United States
| | - Jon A Steingrimsson
- Department of Biostatistics, Brown University, Providence, RI 02903, United States
| |
Collapse
|
15
|
Dinakaran D, Moore-Palhares D, Yang F, Hill JB. Precision radiotherapy with molecular-profiling of CNS tumours. J Neurooncol 2025; 172:51-75. [PMID: 39699761 DOI: 10.1007/s11060-024-04911-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: 08/28/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024]
Abstract
Diagnoses of CNS malignancies in the primary and metastatic setting have significantly advanced in the last decade with the advent of molecular pathology. Using a combination of immunohistochemistry, next-generation sequencing, and methylation profiling integrated with traditional histopathology, patient prognosis and disease characteristics can be understood to a much greater extent. This has recently manifested in predicting response to targeted drug therapies that are redefining management practices of CNS tumours. Radiotherapy, along with surgery, still remains an integral part of treating the majority of CNS tumours. However, the rapid advances in CNS molecular diagnostics have not yet been effectively translated into improving CNS radiotherapy. We explore several promising strategies under development to integrate molecular oncology into radiotherapy, and explore future directions that can serve to use molecular diagnostics to personalize radiotherapy. Evolving the management of CNS tumours with molecular profiling will be integral to supporting the future of precision radiotherapy.
Collapse
Affiliation(s)
- Deepak Dinakaran
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada.
- Department of Medical Biophysics and Radiation Oncology, Temerty Faculty of Medicine, University of Toronto, 149 College Street, Suite 504, Toronto, ON, M5T 1P5, Canada.
| | - Daniel Moore-Palhares
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - Fan Yang
- Radiation Oncology, Mayo Clinic Arizona, 5881 E. Mayo Blvd, Phoenix, AZ, 85054, USA
| | - Jordan B Hill
- Banner MD Anderson Cancer Center, 925 E. McDowell Rd, Phoenix, AZ, 85006, USA
| |
Collapse
|
16
|
Siegel BI, Patil P, Prakash A, Klawinski DM, Hwang EI. Targeted therapy in pediatric central nervous system tumors: a review from the National Pediatric Cancer Foundation. Front Oncol 2025; 15:1504803. [PMID: 40094009 PMCID: PMC11906681 DOI: 10.3389/fonc.2025.1504803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/23/2025] [Indexed: 03/19/2025] Open
Abstract
Central nervous system tumors represent the leading cause of cancer-related mortality in children. Conventional therapies of surgery, radiation, and cytotoxic chemotherapy have insufficient efficacy for some pediatric CNS tumors and are associated with significant morbidity, prompting an ongoing need for novel treatment approaches. Identification of molecular alterations driving tumorigenesis has led to a rising interest in developing targeted therapies for these tumors. The present narrative review focuses on recent progress in targeted therapies for pediatric CNS tumors. We outline the key implicated cellular pathways, discuss candidate molecular therapies for targeting each pathway, and present an overview of the clinical trial landscape for targeted therapies in pediatric CNS tumors. We then discuss challenges and future directions for targeted therapy, including combinatorial approaches and real-time drug screening for personalized treatment planning.
Collapse
Affiliation(s)
- Benjamin I. Siegel
- Brain Tumor Institute and Gilbert Family Neurofibromatosis Institute, Children’s National Hospital, Washington, DC, United States
- Division of Oncology, Children’s National Hospital, Washington, DC, United States
| | - Prabhumallikarjun Patil
- Children’s Healthcare of Atlanta, Aflac Cancer Center, Atlanta, GA, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Akul Prakash
- New York University, New York, NY, United States
| | - Darren M. Klawinski
- Division of Hematology/Oncology, Nemours Children’s Health Jacksonville, Jacksonville, FL, United States
| | - Eugene I. Hwang
- Brain Tumor Institute and Gilbert Family Neurofibromatosis Institute, Children’s National Hospital, Washington, DC, United States
| |
Collapse
|
17
|
Pearson AD, Mueller S, Filbin MG, Grill J, Hawkins C, Jones C, Donoghue M, Drezner N, Weiner S, Russo M, Dun MD, Allen JE, Alonso M, Benaim E, Buenger V, de Rojas T, Desserich K, Fox E, Friend J, Glade Bender J, Hargrave D, Jensen M, Kholmanskikh O, Kieran MW, Knoderer H, Koschmann C, Lesa G, Ligas F, Lipsman N, Ludwinski D, Marshall L, McDonough J, McNicholl AG, Mirsky D, Monje M, Nysom K, Pappo A, Rosenfield A, Scobie N, Slaughter J, Smith M, Souweidane M, Straathof K, Ward L, Weigel B, Zamoryakhin D, Karres D, Vassal G. Paediatric strategy forum for medicinal product development in diffuse midline gliomas in children and adolescents ACCELERATE in collaboration with the European Medicines Agency with participation of the Food and Drug Administration. Eur J Cancer 2025; 217:115230. [PMID: 39854822 DOI: 10.1016/j.ejca.2025.115230] [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: 11/14/2024] [Revised: 01/02/2025] [Accepted: 01/06/2025] [Indexed: 01/27/2025]
Abstract
Fewer than 10 % of children with diffuse midline glioma (DMG) survive 2 years from diagnosis. Radiation therapy remains the cornerstone of treatment and there are no medicinal products with regulatory approval. Although the biology of DMG is better characterized, this has not yet translated into effective treatments. H3K27-alterations initiate the disease but additional drivers are required for malignant growth. Hence, there is an urgent unmet need to develop new multi-modality therapeutic strategies, including alternative methods of drug delivery. ONC201 (DRD2 antagonist and mitochondrial ClpP agonist) is the most widely evaluated investigational drug. Encouraging early data is emerging for CAR T-cells and oncolytic viruses. GD2, B7-H3 and PI3K signalling are ubiquitous targets across all subtypes and therapeutics directed to these targets would potentially benefit the largest number of children. PI3K, ACVR1, MAPK and PDGFRA pathways should be targeted in rational biological combinations. Drug discovery is a very high priority. New specific and potent epigenetic modifiers (PROTACS e.g. SMARCA4 degraders), with blood-brain penetrance are needed. Cancer neuroscience therapeutics are in early development. Overall survival is the preferred regulatory endpoint. However, the evaluation of this can be influenced by the use of re-irradiation at the time of progression. An efficient clinical trial design fit for regulatory purposes for the evaluation of new therapeutics would aid industry and facilitate more efficient therapy development. Challenges in conducting clinical trials such as the need for comparator data and defining endpoints, could be addressed through an international, first-in-child, randomised, complex innovative design trial. To achieve progress: i) drug discovery; ii) new multi-modality, efficient, collaborative, pre-clinical approaches, possibly including artificial intelligence and, iii) efficient clinical trial designs fit for regulatory purposes are required.
Collapse
Affiliation(s)
| | - Sabine Mueller
- Departments of Neurological Surgery, Pediatrics and, Neurology University of California, San Francisco, California, USA. Department of Oncology, University Children's Hospital Zürich, Zürich, Switzerland
| | - Mariella G Filbin
- Broad Institute of Harvard and MIT, Cambridge, MA, USA; Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | | | - Cynthia Hawkins
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Chris Jones
- The Institute of Cancer Research, Sutton, Surrey, UK
| | | | - Nicole Drezner
- US Food and Drug Administration, Silver Springs, MD, USA
| | - Susan Weiner
- ACCELERATE, Europe; Children's Cancer Cause, Washington, DC, USA; Memorial Sloan Kettering Cancer Centre, New York, USA
| | | | - Matthew D Dun
- 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, Mark Hughes Foundation for Brain Cancer Research, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | | | - Marta Alonso
- Program of Solid Tumors, Center for the Applied Medical Research, Pamplona, Spain; Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Center for the Applied Medical Research, Pamplona, Spain
| | | | - Vickie Buenger
- Coalition Against Childhood Cancer (CAC2), Philadelphia, USA
| | | | | | | | | | | | - Darren Hargrave
- University College London Great Ormond Street Institute of Child Health, London, UK
| | | | | | | | | | | | - Giovanni Lesa
- Paediatric Medicines Office, Scientific Evidence Generation Department, Human Division, European Medicines Agency, The Netherlands
| | - Franca Ligas
- Paediatric Medicines Office, Scientific Evidence Generation Department, Human Division, European Medicines Agency, The Netherlands
| | - Nir Lipsman
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | | | - Lynley Marshall
- The Institute of Cancer Research, London, UK; The Royal Marsden Hospital, London, UK
| | | | | | - David Mirsky
- University of Colorado, School of Medicine, CO, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA, Howard Hughes Medical Institute, Stanford, CA, USA
| | | | | | | | | | | | | | | | - Karin Straathof
- University College London Cancer Institute, Great Ormond Street Biomedical Research Centre, London, UK
| | - Lisa Ward
- DIPG-DMG Research Funding Alliance DDRFA /Tough2gether, Manhattan, KS, USA
| | | | | | - Dominik Karres
- Paediatric Medicines Office, Scientific Evidence Generation Department, Human Division, European Medicines Agency, The Netherlands
| | - Gilles Vassal
- ACCELERATE, Europe; Gustave Roussy Cancer Centre, Paris, France
| |
Collapse
|
18
|
Fu Y, Yuan Y, Tan R, Jiang J, Li Z, Li T, Xie G, Xiao Y, Sun H. Development of Small Molecular Hyper-activators of Human Caseinolytic Peptidase P (hClpP) with a [1,8]-naphthyridinone Scaffold as Novel Anti-cancer Agents. ChemMedChem 2025; 20:e202400528. [PMID: 39505831 DOI: 10.1002/cmdc.202400528] [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: 08/15/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024]
Abstract
Based on a clinical staged small molecular hClpP activator ONC201, a class of novel hClpP agonists with a [1,8]naphthyridinone scaffold was designed, synthesized and evaluated in a series of biochemical and biological assays. Mechanism studies for the representative compound F20 indicated that it can potently bind to and activate hClpP, efficiently promote the degradation of hClpP substrates, robustly induce ATF4/CHOP regulated integrated stress responses, strongly inhibit cell growth and effectively induce apoptosis in a subset of cancer cell lines. F20 showed good PK profiles when dosed by intravenous injection and exhibited moderate oral bioavailability in mice.
Collapse
Affiliation(s)
- Yuantao Fu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Yinan Yuan
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Rongliang Tan
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Jinxin Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhilong Li
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Tong Li
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Guangjun Xie
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| | - Yibei Xiao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Haiying Sun
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
19
|
Lemoine C, Da Veiga MA, Rogister B, Piette C, Neirinckx V. An integrated perspective on single-cell and spatial transcriptomic signatures in high-grade gliomas. NPJ Precis Oncol 2025; 9:44. [PMID: 39934275 DOI: 10.1038/s41698-025-00830-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 02/01/2025] [Indexed: 02/13/2025] Open
Abstract
High-grade gliomas (HGG) are incurable brain malignancies in children and adults. Breakthrough advances in transcriptomic technologies unveiled the intricate diversity of cellular states and their spatial organization within HGGs. We qualitatively integrated 55 neoplastic transcriptomic signatures described in 17 single-cell and spatial RNA sequencing-based studies. Our review delineates a spectrum of cellular states, represented by the expression of specific genes, which can be conceptualized along a "reactive-developmental programs" axis. Additionally, we discussed the potential cues influencing these cellular states, including how spatial organization may impact transcriptomic dynamics. Leveraging these insightful discoveries, we discussed a novel, evolutive way to integrate the different transcriptomic signatures in two or three dimensions, incorporating developmental states, their proliferative capacity, and their possible transition towards reactive states. This integrated analysis illuminates the diverse cellular landscape of HGGs and provides a valuable resource for further elucidation of malignant mechanisms, and for the design of therapeutic endeavors.
Collapse
Affiliation(s)
- Célia Lemoine
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000, Liège, Belgium
| | - Marc-Antoine Da Veiga
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000, Liège, Belgium
| | - Bernard Rogister
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000, Liège, Belgium
- Department of Neurology, CHU of Liège, 4000, Liège, Belgium
| | - Caroline Piette
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000, Liège, Belgium
- Department of Pediatrics, Division of Hematology-Oncology, CHU Liège, 4000, Liège, Belgium
| | - Virginie Neirinckx
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000, Liège, Belgium.
| |
Collapse
|
20
|
Sojka C, Wang HLV, Bhatia TN, Li Y, Chopra P, Sing A, Voss A, King A, Wang F, Joseph K, Ravi VM, Olson J, Hoang K, Nduom E, Corces VG, Yao B, Sloan SA. Mapping the developmental trajectory of human astrocytes reveals divergence in glioblastoma. Nat Cell Biol 2025; 27:347-359. [PMID: 39779941 DOI: 10.1038/s41556-024-01583-9] [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: 06/03/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025]
Abstract
Glioblastoma (GBM) is defined by heterogeneous and resilient cell populations that closely reflect neurodevelopmental cell types. Although it is clear that GBM echoes early and immature cell states, identifying the specific developmental programmes disrupted in these tumours has been hindered by a lack of high-resolution trajectories of glial and neuronal lineages. Here we delineate the course of human astrocyte maturation to uncover discrete developmental stages and attributes mirrored by GBM. We generated a transcriptomic and epigenomic map of human astrocyte maturation using cortical organoids maintained in culture for nearly 2 years. Through this approach, we chronicled a multiphase developmental process. Our time course of human astrocyte maturation includes a molecularly distinct intermediate period that serves as a lineage commitment checkpoint upstream of mature quiescence. This intermediate stage acts as a site of developmental deviation separating IDH-wild-type neoplastic astrocyte-lineage cells from quiescent astrocyte populations. Interestingly, IDH1-mutant tumour astrocyte-lineage cells are the exception to this developmental perturbation, where immature properties are suppressed as a result of D-2-hydroxyglutarate oncometabolite exposure. We propose that this defiance is a consequence of IDH1-mutant-associated epigenetic dysregulation, and we identified biased DNA hydroxymethylation (5hmC) in maturation genes as a possible mechanism. Together, this study illustrates a distinct cellular state aberration in GBM astrocyte-lineage cells and presents developmental targets for experimental and therapeutic exploration.
Collapse
Affiliation(s)
- Caitlin Sojka
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Hsiao-Lin V Wang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
- Emory Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Tarun N Bhatia
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Yangping Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Pankaj Chopra
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Anson Sing
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Anna Voss
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Alexia King
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Feng Wang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Kevin Joseph
- Department of Neurosurgery, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Vidhya M Ravi
- Department of Neurosurgery, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jeffrey Olson
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Kimberly Hoang
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Edjah Nduom
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Victor G Corces
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
- Emory Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Bing Yao
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Steven A Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA.
- Emory Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
21
|
Monzer A, Ghamlouche F, Wakimian K, Ballout F, Al Bitar S, Yehya A, Kanso M, Saheb N, Tawil A, Doughan S, Hussein M, Mukherji D, Faraj W, Allen JE, Prabhu VV, Abou-Antoun T, Gali-Muhtasib H, Abou-Kheir W. ONC206, an imipridone derivative, demonstrates anti-colorectal cancer activity against stem/progenitor cells in 3D cell cultures and in patient-derived organoids. Pharmacol Rep 2025; 77:229-246. [PMID: 39551911 DOI: 10.1007/s43440-024-00676-4] [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: 05/29/2024] [Revised: 10/25/2024] [Accepted: 11/05/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) remains one of the most frequently diagnosed and life-threatening malignancies worldwide. CRC's high recurrence rates and drug resistance have been correlated with a subpopulation of dormant slowly dividing cells termed CRC stem cells (CCSCs). Consequently, there is a pressing need to identify novel therapeutics that can effectively and specifically target CCSCs. Imipridones are promising structurally related anticancer molecules that showed efficacy in several solid and hematological preclinical models and phase I/II/III clinical trials. This study mainly aimed to assess the potential anticancer effects of ONC206, an imipridone derivative, on CRC three-dimensional in vitro culture systems using HCT116 and HT29 cells. Importantly, the study aimed at using CRC patient-derived organoids (PDOs) to test the potential therapeutic effect of ONC206. METHODS Two-dimensional cell proliferation, viability, migration, and invasion assays were used to assess the effects of ONC206 on two colorectal cancer cell lines, HCT116 and HT29, in vitro. Immunofluorescence imaging, flow cytometry, and western blot analysis were also performed to investigate the mechanism of action of this drug. Sphere formation assay and CRC PDOs were employed to evaluate the effect of ONC206 on CRC cells in a 3D setting and specifically its potency in targeting the CRC stem/progenitor subpopulation of cells. RESULTS Our results showed that ONC206 was more potent than its parental molecule ONC201 in inhibiting the proliferation and viability of HCT116 and HT29 cells. Moreover, ONC206 significantly reduced the migration and invasion indices of CRC cells. These effects were accompanied by an increase in reactive oxygen species (ROS) production, sub-G1 phase accumulation, and apoptosis in HCT116 and HT29 cells. Furthermore, ONC206 significantly inhibited the 3D colonospheres growth and self-renewal ability of CCSCs more potently than ONC201, which was associated with a decrease in the expression of CSC-related markers. Lastly, ONC206 significantly reduced the growth of organoids derived from CRC patients. CONCLUSION Collectively, our findings demonstrate that ONC206 is an effective anticancer molecule capable of targeting CCSCs, which may represent a novel therapeutic strategy that can overcome CRC resistance and recurrence.
Collapse
Affiliation(s)
- Alissar Monzer
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, 1107-2020, Lebanon
| | - Fatima Ghamlouche
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, 1107-2020, Lebanon
| | - Kevork Wakimian
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, 1107-2020, Lebanon
| | - Farah Ballout
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, 1107-2020, Lebanon
| | - Samar Al Bitar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, 1107-2020, Lebanon
| | - Amani Yehya
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, 1107-2020, Lebanon
| | - Mariam Kanso
- Department of Surgery, American University of Beirut Medical Center, Beirut, 1107-2020, Lebanon
| | - Nour Saheb
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, 1107-2020, Lebanon
| | - Ayman Tawil
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, 1107-2020, Lebanon
| | - Samer Doughan
- Department of Surgery, American University of Beirut Medical Center, Beirut, 1107-2020, Lebanon
| | - Maher Hussein
- Department of Surgery, American University of Beirut Medical Center, Beirut, 1107-2020, Lebanon
| | - Deborah Mukherji
- Department of Internal Medicine, Division of Hematology/Oncology, Faculty of Medicine, American University of Beirut Medical Center, Beirut, 1107-2020, Lebanon
| | - Walid Faraj
- Department of Surgery, American University of Beirut Medical Center, Beirut, 1107-2020, Lebanon
| | | | | | - Tamara Abou-Antoun
- Pediatric Hematology-Oncology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO, 63110-101, USA
| | - Hala Gali-Muhtasib
- Department of Biology, American University of Beirut, Beirut, 1107-2020, Lebanon
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, 1107-2020, Lebanon.
| |
Collapse
|
22
|
Erez N, Furth N, Fedyuk V, Wadden J, Aittaleb R, Adam T, Schwark K, Niculcea M, Miclea M, Mody R, Franson A, Parmar HA, Ibrahim M, Lau B, Eze A, Nourmohammadi N, Fried I, Nazarian J, Ron G, Venneti S, Koschmann C, Shema E. Single-molecule systems for the detection and monitoring of plasma-circulating nucleosomes and oncoproteins in diffuse midline glioma. Cell Rep Med 2025; 6:101918. [PMID: 39809263 PMCID: PMC11866549 DOI: 10.1016/j.xcrm.2024.101918] [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: 11/14/2023] [Revised: 09/12/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025]
Abstract
The analysis of cell-free tumor DNA (ctDNA) and proteins in the blood of patients with cancer potentiates a new generation of non-invasive diagnostic approaches. However, confident detection of tumor-originating markers is challenging, especially in the context of brain tumors, where these analytes in plasma are extremely scarce. Here, we apply a sensitive single-molecule technology to profile multiple histone modifications on individual nucleosomes from the plasma of patients with diffuse midline glioma (DMG). The system reveals epigenetic patterns unique to DMG, significantly differentiating this group of patients from healthy subjects or individuals diagnosed with other cancer types. We further develop a method to directly quantify the tumor-originating oncoproteins, lysine 27 to methionine substitution in histone H3 (H3-K27M) and mutant p53, from <1 mL of plasma, allowing for the accurate molecular classification of patients with DMG. We show that our strategy correlates with MRI and droplet-digital PCR (ddPCR) measurements of ctDNA, highlighting the clinical potential of single-molecule-based, multi-parametric assays for DMG diagnosis and treatment monitoring.
Collapse
Affiliation(s)
- Nir Erez
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Noa Furth
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Vadim Fedyuk
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Jack Wadden
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Rayan Aittaleb
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Tiffany Adam
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Kallen Schwark
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Michael Niculcea
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Madeline Miclea
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Rajen Mody
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Andrea Franson
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Hemant A Parmar
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Mohannad Ibrahim
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Benison Lau
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Augustine Eze
- Center for Genetic Medicine, Children's National Medical Center, Washington, DC, USA
| | - Niku Nourmohammadi
- Center for Genetic Medicine, Children's National Medical Center, Washington, DC, USA
| | - Iris Fried
- Unit of Pediatric Hematology Oncology, The Eisenberg R&D Authority, Shaare Zedek Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Javad Nazarian
- Center for Genetic Medicine, Children's National Medical Center, Washington, DC, USA
| | - Guy Ron
- Racah Institute of Physics, Hebrew University, Jerusalem, Israel
| | - Sriram Venneti
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Carl Koschmann
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA.
| | - Efrat Shema
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
23
|
Lim-Fat MJ, Bennett J, Ostrom Q, Touat M, Franceschi E, Schulte J, Bindra RS, Fangusaro J, Dhall G, Nicholson J, Jackson S, Davidson TB, Calaminus G, Robinson G, Whittle JR, Hau P, Ramaswamy V, Pajtler KW, Rudà R, Foreman NK, Hervey-Jumper SL, Das S, Dirks P, Bi WL, Huang A, Merchant TE, Fouladi M, Aldape K, Van den Bent MJ, Packer RJ, Miller JJ, Reardon DA, Chang SM, Haas-Kogan D, Tabori U, Hawkins C, Monje M, Wen PY, Bouffet E, Yeo KK. Central nervous system tumors in adolescents and young adults: A Society for Neuro-Oncology Consensus Review on diagnosis, management, and future directions. Neuro Oncol 2025; 27:13-32. [PMID: 39441704 PMCID: PMC11726256 DOI: 10.1093/neuonc/noae186] [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: 10/25/2024] Open
Abstract
Adolescents and young adults (AYAs; ages 15-39 years) are a vulnerable population facing challenges in oncological care, including access to specialized care, transition of care, unique tumor biology, and poor representation in clinical trials. Brain tumors are the second most common tumor type in AYA, with malignant brain tumors being the most common cause of cancer-related death. The 2021 WHO Classification for central nervous system (CNS) Tumors highlights the importance of integrated molecular characterization with histologic diagnosis in several tumors relevant to the AYA population. In this position paper from the Society for Neuro-Oncology (SNO), the diagnosis and management of CNS tumors in AYA is reviewed, focusing on the most common tumor types in this population, namely glioma, medulloblastoma, ependymoma, and CNS germ cell tumor. Current challenges and future directions specific to AYA are also highlighted. Finally, possible solutions to address barriers in the care of AYA patients are discussed, emphasizing the need for multidisciplinary and collaborative approaches that span the pediatric and adult paradigms of care, and incorporating advanced molecular testing, targeted therapy, and AYA-centered care.
Collapse
Affiliation(s)
- Mary Jane Lim-Fat
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Ontario, Canada
| | - Julie Bennett
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Quinn Ostrom
- The Preston Robert Tisch Brain Tumor Center, Duke University School of Medicine, Durham, North Carolina, USA
- Central Brain Tumor Registry of the United States, Hinsdale, Illinois, USA
| | - Mehdi Touat
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neuro-oncologie, Paris, France
- Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Enrico Franceschi
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna / AUSL di Bologna, Bologna, Italy
| | - Jessica Schulte
- Neurosciences Department, University of California San Diego, La Jolla, California, USA
| | - Ranjit S Bindra
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jason Fangusaro
- Children’s Healthcare of Atlanta, Emory University, and the Aflac Cancer Center, Atlanta, Georgia, USA
| | - Girish Dhall
- Department of Hematology and Oncology, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - James Nicholson
- Paediatric Oncology, Cambridge University Hospitals and Department of Paediatrics, Cambridge University, UK
| | - Sadhana Jackson
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA
| | - Tom Belle Davidson
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Gabriele Calaminus
- Paediatric Haematology and Oncology, University Hospital Bonn, Bonn, Germany
| | - Giles Robinson
- Department of Oncology, Neurobiology and Brain Tumor Program, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - James R Whittle
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Personalised Oncology Division, WEHI, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Peter Hau
- Department of Neurology and Wilhelm Sander-Neuro-Oncology Unit, Regensburg University Hospital, Regensburg, Germany
| | - Vijay Ramaswamy
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kristian W Pajtler
- Hopp Children’s Cancer Center Heidelberg (KiTZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Division of Pediatric Neuro-oncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Hematology, Oncology and Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Roberta Rudà
- Division of Neuro-Oncology, Department Neuroscience Rita Levi Montalcini, University of Turin and City of Health and Science University Hospital, Turin, Italy
| | - Nicholas K Foreman
- Center for Cancer and Blood Disorders, Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Shawn L Hervey-Jumper
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Sunit Das
- Division of Neurosurgery, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Peter Dirks
- Division of Neurosurgery, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Wenya Linda Bi
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Annie Huang
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Thomas E Merchant
- Department of Radiation Oncology, St Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Maryam Fouladi
- Division of Hematology/Oncology, University of Cincinnati, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kenneth Aldape
- Laboratory of Pathology, National Cancer Institute, Bethesda, Maryland, USA
| | | | - Roger J Packer
- Brain Tumor Institute, Gilbert Family Neurofibromatosis Institute, Center for Neuroscience and Behavioral Medicine, Children’s National Hospital, Washington, District of Columbia, USA
| | - Julie J Miller
- Center for Neuro-Oncology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - David A Reardon
- Center For Neuro-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Susan M Chang
- Division of Neuro-Oncology, Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Daphne Haas-Kogan
- Brigham and Women’s Hospital, Dana-Farber Cancer Institute, Boston Children’s Hospital, Mass General Brigham, Harvard Medical School, Boston, Massachusetts, USA
| | - Uri Tabori
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Cynthia Hawkins
- Department of Pathology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, California, USA
| | - Patrick Y Wen
- Center For Neuro-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Eric Bouffet
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kee Kiat Yeo
- Department of Pediatric Oncology, Dana Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| |
Collapse
|
24
|
O’Halloran K, Crotty EE, Christodoulou E, Leary SE, Miller A, Paulson VA, Lockwood CM, Margol AS, Biegel JA. Targeted detection of sequence variants in cell-free DNA from cerebrospinal fluid in pediatric central nervous system tumors. Front Oncol 2025; 14:1513073. [PMID: 39834946 PMCID: PMC11743934 DOI: 10.3389/fonc.2024.1513073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 11/29/2024] [Indexed: 01/22/2025] Open
Abstract
The emergence of liquid biopsy technologies holds great promise in the cancer setting, including in pediatric central nervous system (CNS) tumors. In contrast to broad lower-depth sequencing, commonly referred to as low pass whole genome sequencing (WGS), targeted platforms with a higher depth of coverage have also been established. Here, we review targeted liquid biopsy techniques with applicability to pediatric CNS tumors. These include polymerase chain reaction (PCR), both droplet digital PCR and reverse transcription-based PCR, Sanger sequencing, and next-generation sequencing approaches that incorporate amplicon- and hybrid capture-based methods. The goal of this paper is to facilitate an understanding of these targeted techniques and provide a context for clinical relevance within disease categories, as well as a discussion on optimizing real-world implementation for pediatric CNS tumors.
Collapse
Affiliation(s)
- Katrina O’Halloran
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Los Angeles, CA, United States
- Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Erin E. Crotty
- Ben Towne Center for Childhood Cancer and Blood Disorders Research and the Department of Pediatrics, Seattle Children’s Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA, United States
| | - Eirini Christodoulou
- Department of Pathology, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, United States
| | - Sarah E. Leary
- Ben Towne Center for Childhood Cancer and Blood Disorders Research and the Department of Pediatrics, Seattle Children’s Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA, United States
| | - Alexandra Miller
- Brain and Spine Tumor Center, Perlmutter Cancer Center, New York University Langone, New York, NY, United States
- Department of Neurology, New York University-Langone Health, New York, NY, United States
| | - Vera A. Paulson
- Genetics and Solid Tumor Laboratory, Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, United States
| | - Christina M. Lockwood
- Genetics and Solid Tumor Laboratory, Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, United States
| | - Ashley S. Margol
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Los Angeles, CA, United States
- Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Jaclyn A. Biegel
- Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
25
|
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
|
26
|
Komza M, Chipuk JE. Mitochondrial metabolism: A moving target in hepatocellular carcinoma therapy. J Cell Physiol 2025; 240:e31441. [PMID: 39324415 PMCID: PMC11732733 DOI: 10.1002/jcp.31441] [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: 05/01/2024] [Revised: 08/21/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024]
Abstract
Mitochondria are pivotal contributors to cancer mechanisms due to their homeostatic and pathological roles in cellular bioenergetics, biosynthesis, metabolism, signaling, and survival. During transformation and tumor initiation, mitochondrial function is often disrupted by oncogenic mutations, leading to a metabolic profile distinct from precursor cells. In this review, we focus on hepatocellular carcinoma, a cancer arising from metabolically robust and nutrient rich hepatocytes, and discuss the mechanistic impact of altered metabolism in this setting. We provide distinctions between normal mitochondrial activity versus disease-related function which yielded therapeutic opportunities, along with highlighting recent preclinical and clinical efforts focused on targeting mitochondrial metabolism. Finally, several novel strategies for exploiting mitochondrial programs to eliminate hepatocellular carcinoma cells in metabolism-specific contexts are presented to integrate these concepts and gain foresight into the future of mitochondria-focused therapeutics.
Collapse
Affiliation(s)
- Monika Komza
- Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jerry Edward Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Icahn School of Medicine at Mount Sinai, The Tisch Cancer Institute, New York, New York, USA
- The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Icahn School of Medicine at Mount Sinai, The Diabetes, Obesity, and Metabolism Institute, New York, New York, USA
| |
Collapse
|
27
|
Wu LJ, Pinho-Schwermann M, Zhou L, Zhang L, Huntington KE, Malpass R, Seyhan AA, Carneiro BA, El-Deiry WS. Synergistic combination therapy with ONC201 or ONC206, and enzalutamide or darolutamide in preclinical studies of castration-resistant prostate cancer. Am J Cancer Res 2024; 14:6012-6036. [PMID: 39803644 PMCID: PMC11711516 DOI: 10.62347/vjmw4904] [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: 08/29/2024] [Accepted: 11/10/2024] [Indexed: 01/16/2025] Open
Abstract
Androgen receptor (AR) signaling is a target in prostate cancer therapy and can be treated with non-steroidal anti-androgens (NSAA) including enzalutamide, and apalutamide for patients with advanced disease. Metastatic castration-resistant prostate cancer (mCPRC) develop resistance becomes refractory to therapy limiting patient overall survival. Darolutamide is a novel next-generation androgen receptor-signaling inhibitor that is FDA approved for non-metastatic castration resistant prostate cancer (nmCRPC). Imipridone ONC201/TIC10 is first-in-class small molecule imipridone that activates the integrated stress response (ISR), upregulates TNF-related apoptosis-inducing ligand (TRAIL) and has activity against CRPC alone or in combination with enzalutamide in preclinical models. We hypothesized that combination of imipridones with androgen receptor signaling blockers such as darolutamide may synergize in anti-tumor efficacy against mCRPC cells. mCRPC cell lines 22RV1, LNCaP, DU145 and PC3 were treated with imipridones ONC201, ONC206, apalutamide, darolutamide, or enzalutamide as single agents or in combinations. Combinations of ONC201 or ONC206 and androgen receptor signaling blockers demonstrated synergistic effects in mCRPC cells. Combinations of ONC201 and darolutamide or enzalutamide reduced PSA levels in LNCaP cells and induced of ATF4 in both LNCaP and 22RV1 cell lines. Darolutamide synergized with ONC201 regardless of AR status or castration sensitivity in vitro. Flow cytometric analysis showed increased intra-tumoral NK cells in mice treated with ONC201 and combination of ONC201 and darolutamide. Trends of increased TRAIL activation within NK cells were also observed in treatment groups. ONC201 and darolutamide demonstrated anti-tumor effects in vivo in the 22RV1 CRPC model. Our results prompt further translational and clinical studies with imipridones ONC201 or ONC206 in combination with enzalutamide or darolutamide for treatment of castrate resistant advanced or metastatic prostate cancer.
Collapse
Affiliation(s)
- Laura Jinxuan Wu
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Biotechnology Graduate Program, Brown UniversityProvidence, RI 02903, USA
| | - Maximilian Pinho-Schwermann
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- The Joint Program in Cancer Biology, Brown University and The Lifespan Health SystemProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Lanlan Zhou
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- The Joint Program in Cancer Biology, Brown University and The Lifespan Health SystemProvidence, RI 02903, USA
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Leiqing Zhang
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Kelsey E Huntington
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Pathobiology Graduate Program, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Ryan Malpass
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Attila A Seyhan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- The Joint Program in Cancer Biology, Brown University and The Lifespan Health SystemProvidence, RI 02903, USA
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Benedito A Carneiro
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- The Joint Program in Cancer Biology, Brown University and The Lifespan Health SystemProvidence, RI 02903, USA
- Hematology-Oncology Division, Department of Medicine, Rhode Island Hospital and Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Wafik S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- The Joint Program in Cancer Biology, Brown University and The Lifespan Health SystemProvidence, RI 02903, USA
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Pathobiology Graduate Program, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Hematology-Oncology Division, Department of Medicine, Rhode Island Hospital and Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| |
Collapse
|
28
|
Chen B, Sun M, Zhang C, Huang Q, Teng D, Hu L, Ma H, Lin X, Huang Z, Gui R, Hu X, Xu L, Zheng M, Zhou Y, Li J, Wang M. Discovery of CLPP-1071 as an Exceptionally Potent and Orally Efficacious Human ClpP Activator with Strong In Vivo Antitumor Activity. J Med Chem 2024; 67:21009-21029. [PMID: 39574384 DOI: 10.1021/acs.jmedchem.4c01605] [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: 12/13/2024]
Abstract
Human sapiens caseinolytic protease P (ClpP) is essential for maintaining mitochondrial proteome homeostasis, and its activation is increasingly recognized as a promising cancer therapy strategy. Herein, based on structure-guided drug design, we discovered a series of potent ClpP activators by introducing a methyl group to the imipridone scaffold of the ClpP activator ONC201 in Phase III clinical trials. Through structural optimization of the lead compound, the most optimal compound, CLPP-1071, exhibited exceptionally potent ClpP agonistic activity (EC50 = 23.5 nM, 107.1-fold stronger than ONC201) and inhibited the proliferation of HL60 cells (IC50 = 4.6 nM, 169.2-fold stronger than ONC201). CLPP-1071 possesses good pharmacokinetic properties and effectively prolongs the lifespan in the MOLM13 and HL60 xenograft models in mice through oral administration. CLPP-1071 is the most potent and orally efficacious ClpP activator reported to date.
Collapse
Affiliation(s)
- Beijing Chen
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan 528400, Guangdong, China
- School of Pharmacy, Guizhou Medical University, Guiyang 550014, China
| | - Mingyang Sun
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan 528400, Guangdong, China
| | - Chun Zhang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan 528400, Guangdong, China
- School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Qi Huang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan 528400, Guangdong, China
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Dan Teng
- Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Linghao Hu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan 528400, Guangdong, China
- Department of Medicinal Chemistry, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Huicong Ma
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan 528400, Guangdong, China
- School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Xinyi Lin
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan 528400, Guangdong, China
- School of Pharmaceutical Sciences, Southern Medical University, No. 1023 South Shatai Road Baiyun District, Guangzhou 510515, Guangdong, China
| | - Zan Huang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan 528400, Guangdong, China
| | - Renzhao Gui
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan 528400, Guangdong, China
| | - Xiaobei Hu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan 528400, Guangdong, China
| | - Lei Xu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan 528400, Guangdong, China
| | - Mingyue Zheng
- Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Yubo Zhou
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan 528400, Guangdong, China
- State Key Laboratory of Drug Research, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Jia Li
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan 528400, Guangdong, China
- State Key Laboratory of Drug Research, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- State Key Laboratory of Chemical Biology, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- School of Pharmacy, Guizhou Medical University, Guiyang 550014, China
| | - Mingliang Wang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Tsuihang New District, Zhongshan 528400, Guangdong, China
- Department of Medicinal Chemistry, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| |
Collapse
|
29
|
Hansford JR, Bouche G, Ramaswamy V, Jabado N, Fonseca A, Moloney S, Gottardo NG, Robinson GW, Gajjar A, Tinkle CL, Fisher PG, Foreman N, Ashley DM, Ziegler DS, Eisenstat DD, Massimino M, Witt O, Bartels U, Rutkowski S, Hargrave D, Fouladi M, Pfister SM, Bouffet E. Comments and Controversies in Oncology: The Tribulations of Trials Developing ONC201. J Clin Oncol 2024; 42:4126-4129. [PMID: 39110925 DOI: 10.1200/jco.24.00709] [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: 04/04/2024] [Revised: 06/09/2024] [Accepted: 07/08/2024] [Indexed: 12/07/2024] Open
Abstract
Our international team highlights issues with efficacy reports in several studies on DMG with the new drug ONC201.
Collapse
Affiliation(s)
- Jordan R Hansford
- Michael Rice Centre for Hematology and Oncology, Women's and Children's Hospital; South Australia Health and Medical Research Institute; South Australia ImmunoGENomics Cancer Institute, University of Adelaide, Adelaide, Australia
| | | | - Vijay Ramaswamy
- Developmental and Stem Cell Biology, Labatt Brain Tumor Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
- Division of Hematology/Oncology, Hospital for Sick Children, Toronto, ON, Canada
- Departments of Medical Biophysics and Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Nada Jabado
- Department of Pediatrics, McGill University, RI-MUHC, Montreal, QC, Canada
| | | | - Sam Moloney
- Children's Cancer Centre, Royal Children's Hospital; Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Nicholas G Gottardo
- Department of Pediatric and Adolescent Oncology and Hematology, Perth Children's Hospital; Telethon Kid's Cancer Centre, Telethon Kid's Institute, University of Western Australia, Perth, WA, Australia
| | | | - Amar Gajjar
- St Jude Children's Research Hospital, Memphis, TN
| | | | | | | | - David M Ashley
- Preston Robert Tisch Brain Tumor Centre, Duke University, Durham, NC
| | - David S Ziegler
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Kensington, NSW, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - David D Eisenstat
- Children's Cancer Centre, Royal Children's Hospital; Murdoch Children's Research Institute; Department of Pediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Maura Massimino
- Pediatric Oncology Unit, Fondazione IRCCS Instituto Nazionale dei Tumori, Milan, Italy
| | - Olaf Witt
- Hopp Children's Cancer Centre Heidelberg (KiTZ), Heidelberg, Germany
- Department of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer 36 Consortium (DKTK), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology & Immunology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Ute Bartels
- Hopp Children's Cancer Centre Heidelberg (KiTZ), Heidelberg, Germany
| | | | - Darren Hargrave
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Maryam Fouladi
- Department of Neuro-Oncology, Nationwide Children's Hospital, Columbus, OH
| | - Stefan M Pfister
- Hopp Children's Cancer Centre Heidelberg (KiTZ), Heidelberg, Germany
| | - Eric Bouffet
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
30
|
Giarrizzo M, LaComb JF, Patel HR, Reddy RG, Haley JD, Graves LM, Iwanowicz EJ, Bialkowska AB. TR-107, an Agonist of Caseinolytic Peptidase Proteolytic Subunit, Disrupts Mitochondrial Metabolism and Inhibits the Growth of Human Colorectal Cancer Cells. Mol Cancer Ther 2024; 23:1761-1778. [PMID: 39233476 PMCID: PMC11614700 DOI: 10.1158/1535-7163.mct-24-0170] [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: 03/14/2024] [Revised: 07/01/2024] [Accepted: 08/22/2024] [Indexed: 09/06/2024]
Abstract
Oxidative phosphorylation is an essential metabolic process for cancer proliferation and therapy resistance. The ClpXP complex maintains mitochondrial proteostasis by degrading misfolded proteins. Madera Therapeutics has developed a class of highly potent and selective small-molecule activators (TR compounds) of the ClpXP component caseinolytic peptidase proteolytic subunit (ClpP). This approach to cancer therapy eliminates substrate recognition and activates nonspecific protease function within mitochondria, which has shown encouraging preclinical efficacy in multiple malignancies. The class-leading compound TR-107 has demonstrated significantly improved potency in ClpP affinity and activation and enhanced pharmacokinetic properties over the multitargeting clinical agent ONC201. In this study, we investigate the in vitro efficacy of TR-107 against human colorectal cancer cells. TR-107 inhibited colorectal cancer cell proliferation in a dose- and time-dependent manner and induced cell cycle arrest at low nanomolar concentrations. Mechanistically, TR-107 downregulated the expression of proteins involved in the mitochondrial unfolded protein response and mitochondrial DNA transcription and translation. TR-107 attenuated oxygen consumption rate and glycolytic compensation, confirming inactivation of oxidative phosphorylation and a reduction in total cellular respiration. Multiomics analysis of treated cells indicated a downregulation of respiratory chain complex subunits and an upregulation of mitophagy and ferroptosis pathways. Further evaluation of ferroptosis revealed a depletion of antioxidant and iron toxicity defenses that could potentiate sensitivity to combinatory chemotherapeutics. Together, this study provides evidence and insight into the subcellular mechanisms employed by colorectal cancer cells in response to potent ClpP agonism. Our findings demonstrate a productive approach to disrupting mitochondrial metabolism, supporting the translational potential of TR-107.
Collapse
Affiliation(s)
- Michael Giarrizzo
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Joseph F LaComb
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Hetvi R Patel
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Rohan G Reddy
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - John D Haley
- Department of Pathology, Stony Brook University, Stony Brook, New York
- Developmental Therapeutics at SBU Cancer Center, Stony Brook University, Stony Brook, New York
- SBU Proteomics Center, Stony Brook University, Stony Brook, New York
| | - Lee M Graves
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | - Agnieszka B Bialkowska
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| |
Collapse
|
31
|
Cassim A, Dun MD, Gallego-Ortega D, Valdes-Mora F. EZHIP's role in diffuse midline glioma: echoes of oncohistones? Trends Cancer 2024; 10:1095-1105. [PMID: 39343635 DOI: 10.1016/j.trecan.2024.09.002] [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: 04/27/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024]
Abstract
The enhancer of zeste inhibitory protein (EZHIP) is typically expressed during germ cell development and has been classified as a cancer-testis antigen (CTA) in various cancers. In 2020, 4% of diffuse midline gliomas (DMGs) were shown to aberrantly express EZHIP, mirroring the DMG hallmark histone H3 K27M (H3K27M) oncohistone mutation. Similar to H3K27M, EZHIP is a negative regulator of polycomb repressive complex 2 (PRC2), leading to global epigenomic remodeling. In this opinion, we explore the similarities and disparities between H3K27M- and EZHIP-DMGs with a focus on their shared functional hallmark of PRC2 inhibition, their genetic and epigenomic landscapes, plausible differences in the cell of origin, and therapeutic avenues. Upcoming research on EZHIP will help better understand its role in gliomagenesis and DMG therapy.
Collapse
Affiliation(s)
- Afraah Cassim
- Cancer Epigenetic Biology and Therapeutics Laboratory, Children's Cancer Institute, Lowy Cancer Centre, Kensington, New South Wales, Australia; School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, 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 Stream, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine, and Wellbeing, Callaghan, New South Wales, Australia
| | - David Gallego-Ortega
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, New South Wales, Australia; School of Clinical Medicine, Faculty of Medicine & Health, University of New South Wales Sydney, New South Wales, Australia; Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Fatima Valdes-Mora
- Cancer Epigenetic Biology and Therapeutics Laboratory, Children's Cancer Institute, Lowy Cancer Centre, Kensington, New South Wales, Australia; School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, New South Wales, Australia; School of Clinical Medicine, Faculty of Medicine & Health, University of New South Wales Sydney, New South Wales, Australia; Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.
| |
Collapse
|
32
|
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
|
33
|
Zhang Y, Jiang J, Ding H, Li Q, Xiao Y, Sun H. Development of novel imipridone derivatives with potent anti-cancer activities as human caseinolytic peptidase P (hClpP) activators. Bioorg Chem 2024; 153:107765. [PMID: 39243740 DOI: 10.1016/j.bioorg.2024.107765] [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: 05/14/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/09/2024]
Abstract
Based on a clinically staged small molecular hClpP activator ONC201, a class of imipridone derivatives was designed and synthesized. These compounds were evaluated in a protease hydrolytic assay, as well as cell growth inhibition assays in three cancer cell lines, MIA PACA-2, HCT116, and MV4-11. A number of compounds that can more potently activate hClpP and more effectively inhibit cell growth in the three cancer cell lines than ONC201 were identified. The most potent compound, ZYZ-17, activated hClpP with an EC50 value of 0.24 µM and inhibited the growth of the three cancer cell lines with IC50 values of less than 10 nM. Mechanism studies for ZYZ-17 revealed that it potently activates cellular hClpP, efficiently induces the degradation of hClpP substrates, and robustly induces apoptosis in the three cancer cell lines. Furthermore, ZYZ-17 demonstrated a promising pharmacokinetic (PK) profile and exhibited highly potent in vivo antitumor activity in a pancreatic cancer MIA PACA-2 xenograft model in BALB/c nude mice.
Collapse
Affiliation(s)
- Yanzhi Zhang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Jinxin Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Hao Ding
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qiannan Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yibei Xiao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Haiying Sun
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
34
|
Hansford JR, Manoharan N, Bouche G, Ramaswamy V, Jabado N, Bouffet E. ONC201 (Dordaviprone): review of evidence to date in diffuse midline glioma, hope or hype? Expert Opin Emerg Drugs 2024; 29:321-325. [PMID: 39548664 DOI: 10.1080/14728214.2024.2426649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 10/30/2024] [Accepted: 10/23/2024] [Indexed: 11/18/2024]
Affiliation(s)
- Jordan R Hansford
- Michael Rice Centre for Hematology and Oncology, Women's and Children's Hospital, Adelaide, SA, Australia
- Paediatric Neuro-Oncology, Cancer Precision Medicine, South Australia Health and Medical Research Institute, Adelaide, SA, Australia
- South Australia ImmunoGENomics Cancer Institute, University of Adelaide, Adelaide, SA, Australia
| | - Neevika Manoharan
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Kensington, NSW, Australia
| | | | - Vijay Ramaswamy
- Developmental and Stem Cell Biology, Labatt Brain Tumor Research Centre, The Hospital for Sick Children, Toronto, Canada
- Division of Hematology/Oncology, Hospital for Sick Children, Toronto, ON, Canada
- Departments of Medical Biophysicsand Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Nada Jabado
- Department of Pediatrics, McGill University, RI-MUHC, Glen Site, Montreal, QC, Canada
| | - Eric Bouffet
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
35
|
Chattopadhyay C, Roszik J, Bhattacharya R, Alauddin M, Mahmud I, Yadugiri S, Ali MM, Khan FS, Prabhu VV, Lorenzi PL, Wei B, Burton E, Morey RR, Lazcano R, Davies MA, Patel SP, Grimm EA. Imipridones inhibit tumor growth and improve survival in an orthotopic liver metastasis mouse model of human uveal melanoma. Br J Cancer 2024; 131:1846-1857. [PMID: 39394450 PMCID: PMC11589887 DOI: 10.1038/s41416-024-02866-6] [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: 03/28/2024] [Revised: 09/11/2024] [Accepted: 09/23/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Uveal melanoma (UM) is a highly aggressive disease with very few treatment options. We previously demonstrated that mUM is characterized by high oxidative phosphorylation (OXPHOS). Here we tested the anti-tumor, signaling and metabolic effects of imipridones, which are CLPP activators, which inhibit OXPHOS indirectly and have demonstrated safety in patients. METHODS We assessed CLPP expression in UM patient samples. We tested the effects of imipridones (ONC201 and ONC212) on the growth, survival, signaling and metabolism of UM cell lines in vitro, and for therapeutic efficacy in vivo in UM liver metastasis models. RESULTS CLPP expression was detected in primary and mUM patient samples. ONC201 and 212 decreased OXPHOS effectors, inhibited cell growth and migration, and induced apoptosis in human UM cell lines in vitro. ONC212 inhibited OXPHOS, increased metabolic stress and apoptotic pathways, inhibited amino acid metabolism, and induced cell death-related lipids. ONC212 also decreased tumor burden and increased survival in vivo in two UM liver metastasis models. CONCLUSIONS Imipridones are a promising strategy for further testing and development in mUM.
Collapse
Affiliation(s)
- Chandrani Chattopadhyay
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Janos Roszik
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rajat Bhattacharya
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Md Alauddin
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Iqbal Mahmud
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Metabolomics Core Facility, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sirisha Yadugiri
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mir Mustafa Ali
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Fatima S Khan
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | | | - Philip L Lorenzi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Metabolomics Core Facility, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Bo Wei
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Metabolomics Core Facility, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Elizabeth Burton
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rohini R Morey
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rossana Lazcano
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sapna P Patel
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Elizabeth A Grimm
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
36
|
Gatto L, Di Nunno V, Ghelardini A, Tosoni A, Bartolini S, Asioli S, Ratti S, Di Stefano AL, Franceschi E. Targeting Mitochondria in Glioma: New Hopes for a Cure. Biomedicines 2024; 12:2730. [PMID: 39767637 PMCID: PMC11727304 DOI: 10.3390/biomedicines12122730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025] Open
Abstract
Drugs targeting mitochondrial energy metabolism are emerging as promising antitumor therapeutics. Glioma treatment is extremely challenging due to the high complexity of the tumor and the high cellular heterogeneity. From a metabolic perspective, glioma cancer cells can be classified into the oxidative metabolic phenotype (mainly depending on mitochondrial respiration for energy production) and glycolytic phenotype or "Warburg effect" (mainly depending on glycolysis). Herein, we reviewed the function of novel bio-active molecules targeting oxidative phosphorylation (OXPHOS), mitochondrial membrane potential and mitochondrial dynamics. These molecules exhibit intriguing preclinical and clinical results and have been proven to be promising candidates to be further developed for glioma therapy. However, despite these initial encouraging results, it is imperative to rigorously assess the side effects of these metabolic drugs, which have a non-negligible toxicity profile.
Collapse
Affiliation(s)
- Lidia Gatto
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| | - Vincenzo Di Nunno
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| | - Anna Ghelardini
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy;
| | - Alicia Tosoni
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| | - Stefania Bartolini
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| | - Sofia Asioli
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, 40126 Bologna, Italy;
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Stefano Ratti
- Cellular Signalling Laboratory, Anatomy Center, Department of Biomedical Sciences (DIBINEM), University of Bologna, 40126 Bologna, Italy;
| | - Anna Luisa Di Stefano
- Division of Neurosurgery, Azienda USL Toscana Nord Ovest, Spedali Riuniti di Livorno, 56121 Livorno, Italy;
- Department of Neurology, Foch Hospital, 92150 Suresnes, France
| | - Enrico Franceschi
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.D.N.); (A.T.); (S.B.); (E.F.)
| |
Collapse
|
37
|
Yuan Y, Li G, Mei S, Hu M, Chu YH, Hsu YC, Li C, Song J, Hu J, Feng D, Xie F, Guan Y, Yue Q, Liu M, Mao Y. Deep mutual learning on hybrid amino acid PET predicts H3K27M mutations in midline gliomas. NPJ Precis Oncol 2024; 8:274. [PMID: 39587279 PMCID: PMC11589770 DOI: 10.1038/s41698-024-00760-1] [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: 02/27/2024] [Accepted: 11/11/2024] [Indexed: 11/27/2024] Open
Abstract
Predicting H3K27M mutation status in midline gliomas non-invasively is of considerable interest, particularly using deep learning with 11C-methionine (MET) and 18F-fluoroethyltyrosine (FET) positron emission tomography (PET). To optimise prediction efficiency, we derived an assistance training (AT) scheme to allow mutual benefits between MET and FET learning to boost the predictability but still only require either PET as inputs for predictions. Our method significantly surpassed conventional convolutional neural network (CNN), radiomics-based, and MR-based methods, achieved an area under the curve (AUC) of 0.9343 for MET, and an AUC of 0.8619 for FET during internal cross-validation (n = 90). The performance remained high in hold-out testing (n = 19) and consecutive testing cohorts (n = 21), with AUCs of 0.9205 and 0.7404. The clinical feasibility of the proposed method was confirmed by the agreements to multi-departmental decisions and outcomes in pathology-uncertain cases. The findings positions our method as a promising tool for aiding treatment decisions in midline glioma.
Collapse
Affiliation(s)
- Yifan Yuan
- Department of Neurosurgery, Huashan Hospital, Fudan University, Neurosurgical Institute of Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Guanglei Li
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Shuhao Mei
- Department of Neurosurgery, Huashan Hospital, Fudan University, Neurosurgical Institute of Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Mingtao Hu
- Shanghai Artificial Intelligence Laboratory, Shanghai, China
| | - Ying-Hua Chu
- MR Collaboration, Siemens Healthineers Ltd., Shanghai, China
| | - Yi-Cheng Hsu
- MR Collaboration, Siemens Healthineers Ltd., Shanghai, China
| | - Chaolin Li
- School of Education, Guangzhou University, Guangzhou, China
| | - Jianping Song
- Department of Neurosurgery, Huashan Hospital, Fudan University, Neurosurgical Institute of Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Department of Neurosurgery, Fudan University Huashan Hospital Fujian Campus, Fujian Medical University, The First Affiliated Hospital Binhai Campus, National Regional Medical Center, Fuzhou, Fujian, China
| | - Jie Hu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Neurosurgical Institute of Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Danyang Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, China
| | - Fang Xie
- National Center for Neurological Disorders, Shanghai, China
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- National Center for Neurological Disorders, Shanghai, China
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qi Yue
- Department of Neurosurgery, Huashan Hospital, Fudan University, Neurosurgical Institute of Fudan University, Shanghai, China.
- National Center for Neurological Disorders, Shanghai, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China.
| | - Mianxin Liu
- Shanghai Artificial Intelligence Laboratory, Shanghai, China.
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Neurosurgical Institute of Fudan University, Shanghai, China.
- National Center for Neurological Disorders, Shanghai, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China.
| |
Collapse
|
38
|
Wang Z, He L, Fan Z, Luo Y. Patenting perspective of modulators of ClpP endopeptidase: 2019-present. Expert Opin Ther Pat 2024; 34:1073-1084. [PMID: 39267345 DOI: 10.1080/13543776.2024.2404233] [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: 06/27/2024] [Revised: 08/14/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
INTRODUCTION ClpP is a highly conserved serine protease that plays a crucial role in maintaining protein homeostasis in both bacterial cells and human mitochondria. Several studies have demonstrated the potential of ClpP as a drug target, with ClpP modulators, including both inhibitors and activators, showing promise in treating a range of conditions such as drug-resistant bacteria, malignant cancers, and fatty liver disease. AREA COVERED This review provides an overview of patents related to ClpP modulators filed over the last five years, detailing their claims and therapeutic applications. The sources of patent information included databases of the European Patent Office, the China Patent Office and the U.S.A. patent Office, while relevant research articles were accessed through PubMed. EXPERT OPINION The number of patents concerning ClpP modulators is on the rise, reflecting advancements in related research. By summarizing and outlining relevant patents, we aim to stimulate further interest among researchers, ultimately leading to the development of effective drugs based on ClpP modulators. The broad spectrum of diseases associated with ClpP dysfunction underscores the potential for ClpP modulators to address a wide range of therapeutic needs.
Collapse
Affiliation(s)
- Zhenyu Wang
- State Key Laboratory of Biotherapy, West China Hospital, West Medical School, Sichuan University, Chengdu, China
| | - Liqing He
- State Key Laboratory of Biotherapy, West China Hospital, West Medical School, Sichuan University, Chengdu, China
| | - Ziheng Fan
- State Key Laboratory of Biotherapy, West China Hospital, West Medical School, Sichuan University, Chengdu, China
| | - Youfu Luo
- State Key Laboratory of Biotherapy, West China Hospital, West Medical School, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Yang Y, Hong Y, Zhao K, Huang M, Li W, Zhang K, Zhao N. Spatial transcriptomics analysis identifies therapeutic targets in diffuse high-grade gliomas. Front Mol Neurosci 2024; 17:1466302. [PMID: 39530009 PMCID: PMC11552449 DOI: 10.3389/fnmol.2024.1466302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/01/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Diffuse high-grade gliomas are the most common malignant adult neuroepithelial tumors in humans and a leading cause of cancer-related death worldwide. The advancement of high throughput transcriptome sequencing technology enables rapid and comprehensive acquisition of transcriptome data from target cells or tissues. This technology aids researchers in understanding and identifying critical therapeutic targets for the prognosis and treatment of diffuse high-grade glioma. Methods Spatial transcriptomics was conducted on two cases of isocitrate dehydrogenase (IDH) wild-type diffuse high-grade glioma (Glio-IDH-wt) and two cases of IDH-mutant diffuse high-grade glioma (Glio-IDH-mut). Gene set enrichment analysis and clustering analysis were employed to pinpoint differentially expressed genes (DEGs) involved in the progression of diffuse high-grade gliomas. The spatial distribution of DEGs in the spatially defined regions of human glioma tissues was overlaid in the t-distributed stochastic neighbor embedding (t-SNE) plots. Results We identified a total of 10,693 DEGs, with 5,677 upregulated and 5,016 downregulated, in spatially defined regions of diffuse high-grade gliomas. Specifically, SPP1, IGFBP2, CALD1, and TMSB4X exhibited high expression in carcinoma regions of both Glio-IDH-wt and Glio-IDH-mut, and 3 upregulated DEGs (SMOC1, APOE, and HIPK2) and 4 upregulated DEGs (PPP1CB, UBA52, S100A6, and CTSB) were only identified in tumor regions of Glio-IDH-wt and Glio-IDH-mut, respectively. Moreover, Kyoto Encyclopedia of Genes and Genomes (KEGG) and gene ontology (GO) enrichment analyses revealed that upregulated DEGs were closely related to PI3K/Akt signaling pathway, virus infection, and cytokine-cytokine receptor interaction. Importantly, the expression of these DEGs was validated using GEPIA databases. Furthermore, the study identified spatial expression patterns of key regulatory genes, including those involved in protein post-translational modification and RNA binding protein-encoding genes, with spatially defined regions of diffuse high-grade glioma. Discussion Spatial transcriptome analysis is one of the breakthroughs in the field of medical biotechnology as this can map the analytes such as RNA information in their physical location in tissue sections. Our findings illuminate previously unexplored spatial expression profiles of key biomarkers in diffuse high-grade glioma, offering novel insight for the development of therapeutic strategies in glioma.
Collapse
Affiliation(s)
- Yongtao Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yingzhou Hong
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Kai Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Minhao Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wenhu Li
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Kui Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ninghui Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
40
|
Mbah NE, Myers AL, Sajjakulnukit P, Chung C, Thompson JK, Hong HS, Giza H, Dang D, Nwosu ZC, Shan M, Sweha SR, Maydan DD, Chen B, Zhang L, Magnuson B, Zhu Z, Radyk M, Lavoie B, Yadav VN, Koo I, Patterson AD, Wahl DR, Franchi L, Agnihotri S, Koschmann CJ, Venneti S, Lyssiotis CA. Therapeutic targeting of differentiation-state dependent metabolic vulnerabilities in diffuse midline glioma. Nat Commun 2024; 15:8983. [PMID: 39419964 PMCID: PMC11487135 DOI: 10.1038/s41467-024-52973-4] [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: 03/01/2022] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
H3K27M diffuse midline gliomas (DMG), including diffuse intrinsic pontine gliomas (DIPG), exhibit cellular heterogeneity comprising less-differentiated oligodendrocyte precursors (OPC)-like stem cells and more differentiated astrocyte (AC)-like cells. Here, we establish in vitro models that recapitulate DMG-OPC-like and AC-like phenotypes and perform transcriptomics, metabolomics, and bioenergetic profiling to identify metabolic programs in the different cellular states. We then define strategies to target metabolic vulnerabilities within specific tumor populations. We show that AC-like cells exhibit a mesenchymal phenotype and are sensitized to ferroptotic cell death. In contrast, OPC-like cells upregulate cholesterol biosynthesis, have diminished mitochondrial oxidative phosphorylation (OXPHOS), and are accordingly more sensitive to statins and OXPHOS inhibitors. Additionally, statins and OXPHOS inhibitors show efficacy and extend survival in preclinical orthotopic models established with stem-like H3K27M DMG cells. Together, this study demonstrates that cellular subtypes within DMGs harbor distinct metabolic vulnerabilities that can be uniquely and selectively targeted for therapeutic gain.
Collapse
Affiliation(s)
- Nneka E Mbah
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Amy L Myers
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Peter Sajjakulnukit
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA
- Graduate Program in Cancer Biology, University of Michigan, Ann Arbor, USA
| | - Chan Chung
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Ann Arbor, USA
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, South Korea
| | | | - Hanna S Hong
- Graduate Program in Immunology, University of Michigan, Ann Arbor, USA
| | - Heather Giza
- Graduate Program in Cancer Biology, University of Michigan, Ann Arbor, USA
| | - Derek Dang
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Ann Arbor, USA
- Graduate Program in Molecular & Cellular Pathology, University of Michigan, Ann Arbor, USA
| | - Zeribe C Nwosu
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Mengrou Shan
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Stefan R Sweha
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Ann Arbor, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, USA
| | - Daniella D Maydan
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Brandon Chen
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, USA
| | - Li Zhang
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Brian Magnuson
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, USA
| | - Zirui Zhu
- Graduate Program in Chemical Biology, University of Michigan, Ann Arbor, USA
| | - Megan Radyk
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Brooke Lavoie
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Viveka Nand Yadav
- The Department of Pediatrics, Children's Mercy Research Institute (CMRI), Kansas, USA
| | - Imhoi Koo
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, USA
| | - Andrew D Patterson
- Department of Biochemistry and Molecular Biology and Department of Veterinary and Biomedical Sciences, the Pennsylvania State University, University Park, USA
| | - Daniel R Wahl
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, USA
| | - Luigi Franchi
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, USA
| | | | - Carl J Koschmann
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, USA
| | - Sriram Venneti
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA.
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Ann Arbor, USA.
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, USA.
- Department of Pathology, University of Michigan Medical School, Ann Arbor, USA.
| | - Costas A Lyssiotis
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA.
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA.
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, USA.
| |
Collapse
|
41
|
Gaggero G. High-grade gliomas: a unique cohort? An overview of a complex and heterogeneous histomolecular classification system. Rep Pract Oncol Radiother 2024; 29:524-526. [PMID: 39895951 PMCID: PMC11785385 DOI: 10.5603/rpor.101990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025] Open
|
42
|
Jiang J, Xie G, Li T, Ding H, Tang R, Luo J, Li Q, Lu W, Xiao Y, Sun H. Discovery of Dehydrogenated Imipridone Derivatives as Activators of Human Caseinolytic Protease P. J Med Chem 2024; 67:15328-15352. [PMID: 39172943 DOI: 10.1021/acs.jmedchem.4c00798] [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: 08/24/2024]
Abstract
Based on the founding member of imipridones, ONC201, a class of dehydrogenated imipridone derivatives was designed, synthesized, and evaluated in a series of biochemical and biological assays as human caseinolytic protease P (hClpP) activators. Mechanism studies for one of the most potent compounds, XT6, indicated that it can potently bind to both recombinant and cellular hClpP, effectively promote the formation of hClpP tetradecamer, efficiently induce the degradation of hClpP substrates, robustly upregulate the expression of ATF4, and strongly inhibit the phosphorylations of AKT and ERK. More importantly, XT6 exhibited a promising pharmacokinetic profile in rats and could penetrate the blood brain barrier. It showed highly potent in vivo antitumor activity in a MIAPACA2 cell line derived pancreatic cancer model in BALB/c nude mice.
Collapse
Affiliation(s)
- Jinxin Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Guangjun Xie
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Tong Li
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Hao Ding
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Rui Tang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Jiajun Luo
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Qiannan Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Wugang Lu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yibei Xiao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Haiying Sun
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
43
|
Cao J, Cao F, Wang C, Jiao Z, You Y, Wang X, Zhao W. ONC206 targeting ClpP induces mitochondrial dysfunction and protective autophagy in hepatocellular carcinoma cells. Neoplasia 2024; 55:101015. [PMID: 38944913 PMCID: PMC11267062 DOI: 10.1016/j.neo.2024.101015] [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: 12/14/2023] [Revised: 05/30/2024] [Accepted: 06/06/2024] [Indexed: 07/02/2024]
Abstract
Hepatocellular carcinoma (HCC) is the most common form of liver cancer, accounting for approximately 90 % of all cases. ONC201, a member of the imipridone drug family, has shown promising therapeutic potential and a good safety profile in both malignant pediatric central nervous system tumors (diffuse midline glioma [DMG]) and hematologic malignancies. ONC206 is a more potent analog of ONC201. However, the ONC206 potential and mechanism of action in HCC remain to be elucidated. We found that ONC206 hindered HCC growth by suppressing cell proliferation and inducing apoptosis. Moreover, ONC206 induced cytoprotective autophagy, and blocking autophagy enhanced the proapoptotic effect of ONC206. Additionally, ONC206 induced mitochondrial swelling, reduced the mitochondrial membrane potential (MMP), and led to the accumulation of mitochondrial ROS in HCC cells, ultimately resulting in mitochondrial dysfunction. The HCC patient samples exhibited notably elevated levels of caseinolytic protease proteolytic subunit (ClpP), which serves as a mediator of ONC206-induced mitochondrial dysfunction and the activation of protective autophagy. knockdown of ClpP reversed the cytotoxic effects of ONC206 on HCC cells. In summary, our results provide the first insight into the mechanism by which ONC206 exerts its anti-HCC effects and induces protective autophagy in HCC cells through ClpP.
Collapse
Affiliation(s)
- Jiahao Cao
- Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361004, PR China; Department of Thyroid Head and Neck Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, 314099, PR China
| | - Fei Cao
- Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361004, PR China
| | - Chuanzheng Wang
- Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361004, PR China
| | - Zhen Jiao
- Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361004, PR China
| | - Yuting You
- Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361004, PR China
| | - Xiaomin Wang
- Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361004, PR China.
| | - Wenxiu Zhao
- Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361004, PR China.
| |
Collapse
|
44
|
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
|
45
|
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
|
46
|
Ryba A, Özdemir Z, Nissimov N, Hönikl L, Neidert N, Jakobs M, Kalasauskas D, Krigers A, Thomé C, Freyschlag C, Ringel F, Unterberg A, Dao Trong P, Beck J, Heiland D, Meyer B, Vajkoczy P, Onken J, Stummer W, Suero Molina E, Gempt J, Westphal M, Schüller U, Mohme M. Insights from a multicenter study on adult H3 K27M-mutated glioma: Surgical resection's limited influence on overall survival, ATRX as molecular prognosticator. Neuro Oncol 2024; 26:1479-1493. [PMID: 38507506 PMCID: PMC11300017 DOI: 10.1093/neuonc/noae061] [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: 11/04/2023] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND H3 K27M-mutated gliomas were first described as a new grade 4 entity in the 2016 World Health Organization classification. Current studies have focused on its typical appearance in children and young adults, increasing the need to better understand the prognostic factors and impact of surgery on adults. Here, we report a multicentric study of this entity in adults. METHODS We included molecularly confirmed H3 K27M-mutated glioma cases in patients ≥ 18 years diagnosed between 2016 and 2022. Clinical, radiological, and surgical features were analyzed. Univariate and multivariate analyses were performed to identify prognostic factors. RESULTS Among 70 patients with a mean age of 36.1 years, the median overall survival (OS) was 13.6 ± 14 months. Gross-total resection was achieved in 14.3% of patients, whereas 30% had a subtotal resection and 54.3% a biopsy. Tumors located in telencephalon/diencephalon/myelencephalon were associated with a poorer OS, while a location in the mesencephalon/metencephalon showed a significantly longer OS (8.7 vs. 25.0 months, P = .007). Preoperative Karnofsky-Performance Score (KPS) ≤ 80 showed a reduced OS (4.2 vs. 18 months, P = .02). Furthermore, ATRX loss, found in 25.7%, was independently associated with an increased OS (31 vs. 8.3 months, P = .0029). Notably, patients undergoing resection showed no survival benefit over biopsy (12 vs. 11 months, P = .4006). CONCLUSIONS The present study describes surgical features of H3 K27M-mutated glioma in adulthood in a large multicentric study. Our data reveal that ATRX status, location and KPS significantly impact OS in H3 K27M-mutated glioma. Importantly, our dataset indicates that resection does not offer a survival advantage over biopsy.
Collapse
Affiliation(s)
- Alice Ryba
- Department of Neurosurgery, Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Zeynep Özdemir
- Department of Neurosurgery, University Hospital of Münster, Münster, Germany
| | - Nitzan Nissimov
- Department of Neurosurgery, Charité University Hospital Berlin, Berlin, Germany
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
| | - Lisa Hönikl
- Department of Neurosurgery, Technical University Munich, Munich, Germany
| | - Nicolas Neidert
- Department of Neurosurgery, Charité University Hospital Berlin, Berlin, Germany
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
| | - Martin Jakobs
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
- Division of Stereotactic Neurosurgery, Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg University, Medical Faculty, Heidelberg, Germany
| | - Darius Kalasauskas
- Department of Neurosurgery, University Medical Center Mainz, Mainz, Germany
| | - Aleksandrs Krigers
- Department of Neurosurgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Claudius Thomé
- Department of Neurosurgery, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Florian Ringel
- Department of Neurosurgery, University Medical Center Mainz, Mainz, Germany
| | - Andreas Unterberg
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg University, Medical Faculty, Heidelberg, Germany
| | - Philip Dao Trong
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg University, Medical Faculty, Heidelberg, Germany
| | - Jürgen Beck
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
| | - Dieter Henrik Heiland
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
| | - Bernhard Meyer
- Department of Neurosurgery, Technical University Munich, Munich, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité University Hospital Berlin, Berlin, Germany
| | - Julia Onken
- Department of Neurosurgery, Charité University Hospital Berlin, Berlin, Germany
| | - Walter Stummer
- Department of Neurosurgery, University Hospital of Münster, Münster, Germany
| | - Eric Suero Molina
- Department of Neurosurgery, University Hospital of Münster, Münster, Germany
| | - Jens Gempt
- Department of Neurosurgery, Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manfred Westphal
- Department of Neurosurgery, Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrich Schüller
- Institute of Neuropathology, Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children’s Cancer Center Hamburg, Hamburg, Germany
| | - Malte Mohme
- Department of Neurosurgery, Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
47
|
Liu S, Sui J, Luo B, Zhang J, Xiang X, Yang T, Luo Y, Liu J. Discovery of 5-(Piperidin-4-yl)-1,2,4-oxadiazole Derivatives as a New Class of Human Caseinolytic Protease P Agonists for the Treatment of Hepatocellular Carcinoma. J Med Chem 2024; 67:10622-10642. [PMID: 38905539 DOI: 10.1021/acs.jmedchem.4c00080] [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: 06/23/2024]
Abstract
Chemical agonism of human caseinolytic protease P (HsClpP) is increasingly being recognized as a potential anticancer strategy due to its critical role in maintaining mitochondrial homeostasis. We unveil the discovery of 5-(piperidin-4-yl)-1,2,4-oxadiazole derivatives as a novel class of HsClpP agonists and demonstrate for the first time the application of HsClpP agonists in the treatment of hepatocellular carcinoma (HCC) (Pace, A.; Pierro, P. The new era of 1,2,4-oxadiazoles. Org. Biomol. Chem. 2009, 7 (21), 4337-4348). Compound SL44 exhibited potent HsClpP agonistic activity in the α-casein hydrolysis assay (EC50 = 1.30 μM) and inhibited the proliferation of HCCLM3 cells (IC50 = 3.1 μM, 21.4-fold higher than hit ADX-47273). Mechanistically, SL44 induces degradation of respiratory chain complex subunits and leads to apoptosis in HCC cells. In vivo results demonstrated that SL44 has potent tumor growth inhibitory activity and has a superior safety profile compared to the kinase inhibitor sorafenib. Overall, we developed a novel class of HsClpP agonists that can potentially be used for the treatment of HCC.
Collapse
Affiliation(s)
- Song Liu
- Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Sui
- Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Baozhu Luo
- Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiangnan Zhang
- Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinrong Xiang
- Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Yang
- Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Youfu Luo
- Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Liu
- Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
48
|
Monje M, Mahdi J, Majzner R, Yeom K, Schultz LM, Richards RM, Barsan V, Song KW, Kamens J, Baggott C, Kunicki M, Lim AS, Reschke A, Mavroukakis S, Egeler E, Moon J, Patel S, Chinnasamy H, Erickson C, Jacobs A, Duh AK, Rietberg SP, Tunuguntla R, Klysz DD, Fowler C, Green S, Beebe B, Carr C, Fujimoto M, Brown AK, Petersen ALG, McIntyre C, Siddiqui A, Lepori-Bui N, Villar K, Pham K, Bove R, Musa E, Reynolds W, Kuo A, Prabhu S, Rasmussen L, Cornell TT, Partap S, Fisher PG, Campen CJ, Grant G, Prolo L, Ye X, Sahaf B, Davis KL, Feldman SA, Ramakrishna S, Mackall C. Sequential intravenous and intracerebroventricular GD2-CAR T-cell therapy for H3K27M-mutated diffuse midline gliomas. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.25.24309146. [PMID: 38978673 PMCID: PMC11230330 DOI: 10.1101/2024.06.25.24309146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
H3K27M-mutant diffuse midline gliomas (DMGs) express high levels of the GD2 disialoganglioside and chimeric antigen receptor modified T-cells targeting GD2 (GD2-CART) eradicate DMGs in preclinical models. Arm A of the Phase I trial NCT04196413 administered one IV dose of autologous GD2-CART to patients with H3K27M-mutant pontine (DIPG) or spinal (sDMG) diffuse midline glioma at two dose levels (DL1=1e6/kg; DL2=3e6/kg) following lymphodepleting (LD) chemotherapy. Patients with clinical or imaging benefit were eligible for subsequent intracerebroventricular (ICV) GD2-CART infusions (10-30e6 GD2-CART). Primary objectives were manufacturing feasibility, tolerability, and identification of a maximally tolerated dose of IV GD2-CART. Secondary objectives included preliminary assessments of benefit. Thirteen patients enrolled and 11 received IV GD2-CART on study [n=3 DL1(3 DIPG); n=8 DL2(6 DIPG/2 sDMG). GD2-CART manufacturing was successful for all patients. No dose-limiting toxicities (DLTs) occurred on DL1, but three patients experienced DLT on DL2 due to grade 4 cytokine release syndrome (CRS). Nine patients received ICV infusions, which were not associated with DLTs. All patients exhibited tumor inflammation-associated neurotoxicity (TIAN). Four patients demonstrated major volumetric tumor reductions (52%, 54%, 91% and 100%). One patient exhibited a complete response ongoing for >30 months since enrollment. Eight patients demonstrated neurological benefit based upon a protocol-directed Clinical Improvement Score. Sequential IV followed by ICV GD2-CART induced tumor regressions and neurological improvements in patients with DIPG and sDMG. DL1 was established as the maximally tolerated IV GD2-CART dose. Neurotoxicity was safely managed with intensive monitoring and close adherence to a management algorithm.
Collapse
|
49
|
Li YA, Zhao C, Ge JJ, Li C, Xue FJ, Qi SP, Zhao C, Kong CC, Zhang JP. Apatinib combined with temozolomide in diffuse midline glioma: a novel and effective therapy. BMC Cancer 2024; 24:754. [PMID: 38907215 PMCID: PMC11193221 DOI: 10.1186/s12885-024-12373-9] [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] [Accepted: 05/10/2024] [Indexed: 06/23/2024] Open
Abstract
PURPOSE Diffuse midline glioma (DMG), H3 K27M-mutant is a type of diffuse high-grade glioma that occurs in the brain midline carrying an extremely poor prognosis under the best efforts of surgery, radiation, and other therapies. For better therapy, we explored the efficacy and toxicity of a novel therapy that combines apatinib and temozolomide in DMG. METHODS A retrospective analysis of 32 patients with DMG who underwent apatinib plus temozolomide treatment was performed. Apatinib was given 500 mg in adults, 250 mg in pediatric patients once daily. Temozolomide was administered at 200 mg/m2/d according to the standard 5/28 days regimen. The main clinical data included basic information of patients, radiological and pathological characteristics of tumors, treatment, adverse reactions, prognosis. RESULTS The objective response rate was 24.1%, and the disease control rate was 79.3%. The median PFS of all patients was 5.8 months, and median OS was 10.3 months. A total of 236 cycles of treatment were available for safety assessment and the toxicity of the combination therapy was relatively well tolerated. The most common grade 3 toxicities were myelosuppression including leukopenia (5.08%), neutropenia (4.24%), lymphopenia (2.12%), thrombocytopenia (1.69%) and anemia (1.27%). Grade 4 toxicities included neutropenia (2.12%), thrombocytopenia (2.12%) and proteinuria (1.69%). All the adverse events were relieved after symptomatic treatment or dose reduction. CONCLUSIONS Apatinib plus temozolomide could be an effective regimen with manageable toxicities and favorable efficacy and may outperform temozolomide monotherapy, particularly in newly diagnosed adults with tumors located outside the pons. The novel therapy deserves further investigation in adult DMG patients.
Collapse
Affiliation(s)
- Yu-An Li
- Department of Neuro-Oncology, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Chuan Zhao
- Department of Neuro-Oncology, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Jing-Jing Ge
- Department of Neuro-Oncology, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Cheng Li
- Department of Neuro-Oncology, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Feng-Jun Xue
- Department of Neuro-Oncology, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Shao-Pei Qi
- Department of Neuro-Oncology, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Chi Zhao
- Department of Neuro-Oncology, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Chen-Chen Kong
- Department of Neuro-Oncology, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Jun-Ping Zhang
- Department of Neuro-Oncology, Sanbo Brain Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
50
|
Arrillaga-Romany I, Miller JJ. Demonstrated efficacy and mechanisms of sensitivity of ONC201: H3K27M-mutant diffuse midline glioma in the spotlight. Neuro Oncol 2024; 26:991-992. [PMID: 38588460 PMCID: PMC11145450 DOI: 10.1093/neuonc/noae051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024] Open
Affiliation(s)
- Isabel Arrillaga-Romany
- Center for Neuro-Oncology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Julie J Miller
- Center for Neuro-Oncology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Translational Neuro-Oncology Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|