1
|
Garcia-Moure M, Laspidea V, Gupta S, Gillard AG, Khatua S, Parthasarathy A, He J, Lang FF, Fueyo J, Alonso MM, Gomez-Manzano C. The emerging field of viroimmunotherapy for pediatric brain tumors. Neuro Oncol 2024; 26:1981-1993. [PMID: 39148489 PMCID: PMC11534321 DOI: 10.1093/neuonc/noae160] [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: 05/05/2024] [Indexed: 08/17/2024] Open
Abstract
Pediatric brain tumors are the most common solid tumors in children. Even to date, with the advances in multimodality therapeutic management, survival outcomes remain dismal in some types of tumors, such as pediatric-type diffuse high-grade gliomas or central nervous system embryonal tumors. Failure to understand the complex molecular heterogeneity and the elusive tumor and microenvironment interplay continues to undermine therapeutic efficacy. Developing a strategy that would improve survival for these fatal tumors remains unmet in pediatric neuro-oncology. Oncolytic viruses (OVs) are emerging as a feasible, safe, and promising therapy for brain tumors. The new paradigm in virotherapy implies that the direct cytopathic effect is followed, under certain circumstances, by an antitumor immune response responsible for the partial or complete debulking of the tumor mass. OVs alone or combined with other therapeutic modalities have been primarily used in adult neuro-oncology. A surge in encouraging preclinical studies in pediatric brain tumor models recently led to the clinical translation of OVs with encouraging results in these tumors. In this review, we summarize the different virotherapy tested in preclinical and clinical studies in pediatric brain tumors, and we discuss the limitations and future avenues necessary to improve the response of these tumors to this type of therapy.
Collapse
Affiliation(s)
- Marc Garcia-Moure
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Virginia Laspidea
- Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain
| | - Sumit Gupta
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Andrew G Gillard
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Soumen Khatua
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Akhila Parthasarathy
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jiasen He
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Juan Fueyo
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Marta M Alonso
- Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain
- Program of Solid Tumors, Center for the Applied Medical Research, Pamplona, Spain
| | - Candelaria Gomez-Manzano
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
2
|
Mishra DK, Popovski D, Morris SM, Bondoc A, Senthil Kumar S, Girard EJ, Rutka J, Fouladi M, Huang A, Olson JM, Drissi R. Preclinical pediatric brain tumor models for immunotherapy: Hurdles and a way forward. Neuro Oncol 2024; 26:226-235. [PMID: 37713135 PMCID: PMC10836771 DOI: 10.1093/neuonc/noad170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Indexed: 09/16/2023] Open
Abstract
Brain tumors are the most common solid tumor in children and the leading cause of cancer-related deaths. Over the last few years, improvements have been made in the diagnosis and treatment of children with Central Nervous System tumors. Unfortunately, for many patients with high-grade tumors, the overall prognosis remains poor. Lower survival rates are partly attributed to the lack of efficacious therapies. The advent and success of immune checkpoint inhibitors (ICIs) in adults have sparked interest in investigating the utility of these therapies alone or in combination with other drug treatments in pediatric patients. However, to achieve improved clinical outcomes, the establishment and selection of relevant and robust preclinical pediatric high-grade brain tumor models is imperative. Here, we review the information that influenced our model selection as we embarked on an international collaborative study to test ICIs in combination with epigenetic modifying agents to enhance adaptive immunity to treat pediatric brain tumors. We also share challenges that we faced and potential solutions.
Collapse
Affiliation(s)
- Deepak Kumar Mishra
- Center for Childhood Cancer Research, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Dean Popovski
- Arthur and Sonia Labatt Brain Tumor Research Centre, Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Onatario, Canada
| | - Shelli M Morris
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Andrew Bondoc
- Arthur and Sonia Labatt Brain Tumor Research Centre, Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Onatario, Canada
| | - Shiva Senthil Kumar
- Center for Childhood Cancer Research, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Emily J Girard
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - James Rutka
- Division of Neurosurgery, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Maryam Fouladi
- Pediatric Neuro-Oncology Program, Nationwide Children’s Hospital, Columbus, Ohio, USA
- The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Annie Huang
- Arthur and Sonia Labatt Brain Tumor Research Centre, Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Onatario, Canada
| | - James M Olson
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Rachid Drissi
- Center for Childhood Cancer Research, Nationwide Children’s Hospital, Columbus, Ohio, USA
- The Ohio State University College of Medicine, Columbus, Ohio, USA
| |
Collapse
|
3
|
Hartley R, Phoenix TN. MYC Promotes Aggressive Growth and Metastasis of a WNT-Medulloblastoma Mouse Model. Dev Neurosci 2023; 46:167-178. [PMID: 37544301 DOI: 10.1159/000533270] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/24/2023] [Indexed: 08/08/2023] Open
Abstract
Medulloblastoma (MB), the most common malignant pediatric brain tumor, comprises four molecularly and clinically distinct subgroups (termed WNT, SHH, group 3, and group 4). Prognosis varies based on genetic and pathological features associated with each molecular subgroup. WNT-MB, considered low-risk, is rarely metastatic and contains activating mutations in CTNNB1; group 3-MB (GRP3-MB), commonly classified as high-risk, is frequently metastatic and can contain genomic alterations, resulting in elevated MYC expression. Here, we compare model systems of low-risk WNT-MB and high-risk GRP3-MB to identify tumor and microenvironment interactions that could contribute to features associated with prognosis. Compared to GRP3-MB, we find that WNT-MB is enriched in gene sets related to extracellular matrix (ECM) regulation and cellular adhesion. Exogenous expression of MycT58A in a murine WNT-MB model significantly accelerates growth and results in metastatic disease. In addition to decreased ECM regulation and cell adhesion pathways, we also identified immune system interactions among the top downregulated signaling pathways following MycT58A expression. Taken together, our data provide evidence that increased Myc signaling can promote the growth and metastasis in a murine model of WNT-MB.
Collapse
Affiliation(s)
- Rachel Hartley
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio, USA
| | - Timothy N Phoenix
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio, USA
- Research in Patient Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
4
|
McNicholas M, De Cola A, Bashardanesh Z, Foss A, Lloyd CB, Hébert S, Faury D, Andrade AF, Jabado N, Kleinman CL, Pathania M. A Compendium of Syngeneic, Transplantable Pediatric High-Grade Glioma Models Reveals Subtype-Specific Therapeutic Vulnerabilities. Cancer Discov 2023; 13:1592-1615. [PMID: 37011011 PMCID: PMC10326601 DOI: 10.1158/2159-8290.cd-23-0004] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/20/2023] [Accepted: 03/29/2023] [Indexed: 04/04/2023]
Abstract
Pediatric high-grade gliomas (pHGG) are lethal, incurable brain tumors frequently driven by clonal mutations in histone genes. They often harbor a range of additional genetic alterations that correlate with different ages, anatomic locations, and tumor subtypes. We developed models representing 16 pHGG subtypes driven by different combinations of alterations targeted to specific brain regions. Tumors developed with varying latencies and cell lines derived from these models engrafted in syngeneic, immunocompetent mice with high penetrance. Targeted drug screening revealed unexpected selective vulnerabilities-H3.3G34R/PDGFRAC235Y to FGFR inhibition, H3.3K27M/PDGFRAWT to PDGFRA inhibition, and H3.3K27M/PDGFRAWT and H3.3K27M/PPM1DΔC/PIK3CAE545K to combined inhibition of MEK and PIK3CA. Moreover, H3.3K27M tumors with PIK3CA, NF1, and FGFR1 mutations were more invasive and harbored distinct additional phenotypes, such as exophytic spread, cranial nerve invasion, and spinal dissemination. Collectively, these models reveal that different partner alterations produce distinct effects on pHGG cellular composition, latency, invasiveness, and treatment sensitivity. SIGNIFICANCE Histone-mutant pediatric gliomas are a highly heterogeneous tumor entity. Different histone mutations correlate with different ages of onset, survival outcomes, brain regions, and partner alterations. We have developed models of histone-mutant gliomas that reflect this anatomic and genetic heterogeneity and provide evidence of subtype-specific biology and therapeutic targeting. See related commentary by Lubanszky and Hawkins, p. 1516. This article is highlighted in the In This Issue feature, p. 1501.
Collapse
Affiliation(s)
- Michael McNicholas
- Department of Oncology and Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- CRUK Children's Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, United Kingdom
| | - Antonella De Cola
- Department of Oncology and Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- CRUK Children's Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, United Kingdom
| | - Zahedeh Bashardanesh
- Lady Davis Research Institute, Jewish General Hospital, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Amelia Foss
- Department of Oncology and Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- CRUK Children's Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, United Kingdom
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Cameron B. Lloyd
- Department of Oncology and Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- CRUK Children's Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, United Kingdom
| | - Steven Hébert
- Lady Davis Research Institute, Jewish General Hospital, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Damien Faury
- Department of Pediatrics, McGill University, and The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | | | - Nada Jabado
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- Department of Pediatrics, McGill University, and The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Claudia L. Kleinman
- Lady Davis Research Institute, Jewish General Hospital, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Manav Pathania
- Department of Oncology and Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- CRUK Children's Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
5
|
Foss A, Pathania M. Pediatric Glioma Models Provide Insights into Tumor Development and Future Therapeutic Strategies. Dev Neurosci 2023; 46:22-43. [PMID: 37231843 DOI: 10.1159/000531040] [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: 02/20/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
In depth study of pediatric gliomas has been hampered due to difficulties in accessing patient tissue and a lack of clinically representative tumor models. Over the last decade, however, profiling of carefully curated cohorts of pediatric tumors has identified genetic drivers that molecularly segregate pediatric gliomas from adult gliomas. This information has inspired the development of a new set of powerful in vitro and in vivo tumor models that can aid in identifying pediatric-specific oncogenic mechanisms and tumor microenvironment interactions. Single-cell analyses of both human tumors and these newly developed models have revealed that pediatric gliomas arise from spatiotemporally discrete neural progenitor populations in which developmental programs have become dysregulated. Pediatric high-grade gliomas also harbor distinct sets of co-segregating genetic and epigenetic alterations, often accompanied by unique features within the tumor microenvironment. The development of these novel tools and data resources has led to insights into the biology and heterogeneity of these tumors, including identification of distinctive sets of driver mutations, developmentally restricted cells of origin, recognizable patterns of tumor progression, characteristic immune environments, and tumor hijacking of normal microenvironmental and neural programs. As concerted efforts have broadened our understanding of these tumors, new therapeutic vulnerabilities have been identified, and for the first time, promising new strategies are being evaluated in the preclinical and clinical settings. Even so, dedicated and sustained collaborative efforts are necessary to refine our knowledge and bring these new strategies into general clinical use. In this review, we will discuss the range of currently available glioma models, the way in which they have each contributed to recent developments in the field, their benefits and drawbacks for addressing specific research questions, and their future utility in advancing biological understanding and treatment of pediatric glioma.
Collapse
Affiliation(s)
- Amelia Foss
- Department of Oncology and the Milner Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- CRUK Children's Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, UK
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Manav Pathania
- Department of Oncology and the Milner Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- CRUK Children's Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, UK
| |
Collapse
|
6
|
Murdaugh RL, Anastas JN. Applying single cell multi-omic analyses to understand treatment resistance in pediatric high grade glioma. Front Pharmacol 2023; 14:1002296. [PMID: 37205910 PMCID: PMC10191214 DOI: 10.3389/fphar.2023.1002296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 04/20/2023] [Indexed: 05/21/2023] Open
Abstract
Despite improvements in cancer patient outcomes seen in the past decade, tumor resistance to therapy remains a major impediment to achieving durable clinical responses. Intratumoral heterogeneity related to genetic, epigenetic, transcriptomic, proteomic, and metabolic differences between individual cancer cells has emerged as a driver of therapeutic resistance. This cell to cell heterogeneity can be assessed using single cell profiling technologies that enable the identification of tumor cell clones that exhibit similar defining features like specific mutations or patterns of DNA methylation. Single cell profiling of tumors before and after treatment can generate new insights into the cancer cell characteristics that confer therapeutic resistance by identifying intrinsically resistant sub-populations that survive treatment and by describing new cellular features that emerge post-treatment due to tumor cell evolution. Integrative, single cell analytical approaches have already proven advantageous in studies characterizing treatment-resistant clones in cancers where pre- and post-treatment patient samples are readily available, such as leukemia. In contrast, little is known about other cancer subtypes like pediatric high grade glioma, a class of heterogeneous, malignant brain tumors in children that rapidly develop resistance to multiple therapeutic modalities, including chemotherapy, immunotherapy, and radiation. Leveraging single cell multi-omic technologies to analyze naïve and therapy-resistant glioma may lead to the discovery of novel strategies to overcome treatment resistance in brain tumors with dismal clinical outcomes. In this review, we explore the potential for single cell multi-omic analyses to reveal mechanisms of glioma resistance to therapy and discuss opportunities to apply these approaches to improve long-term therapeutic response in pediatric high grade glioma and other brain tumors with limited treatment options.
Collapse
Affiliation(s)
- Rebecca L. Murdaugh
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, United States
- Program in Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Jamie N. Anastas
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, United States
- Program in Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
7
|
Day CA, Hinchcliffe EH, Robinson JP. H3K27me3 in Diffuse Midline Glioma and Epithelial Ovarian Cancer: Opposing Epigenetic Changes Leading to the Same Poor Outcomes. Cells 2022; 11:cells11213376. [PMID: 36359771 PMCID: PMC9655269 DOI: 10.3390/cells11213376] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/19/2022] [Accepted: 10/24/2022] [Indexed: 11/29/2022] Open
Abstract
Histone post-translational modifications modulate gene expression through epigenetic gene regulation. The core histone H3 family members, H3.1, H3.2, and H3.3, play a central role in epigenetics. H3 histones can acquire many post-translational modifications, including the trimethylation of H3K27 (H3K27me3), which represses transcription. Triple methylation of H3K27 is performed by the histone methyltransferase Enhancer of Zeste Homologue 2 (EZH2), a component of the Polycomb Repressive Complex 2. Both global increases and decreases in H3K27me3 have been implicated in a wide range of cancer types. Here, we explore how opposing changes in H3K27me3 contribute to cancer by highlighting its role in two vastly different cancer types; (1) a form of glioma known as diffuse midline glioma H3K27-altered and (2) epithelial ovarian cancer. These two cancers vary widely in the age of onset, sex, associated mutations, and cell and organ type. However, both diffuse midline glioma and ovarian cancer have dysregulation of H3K27 methylation, triggering changes to the cancer cell transcriptome. In diffuse midline glioma, the loss of H3K27 methylation is a primary driving factor in tumorigenesis that promotes glial cell stemness and silences tumor suppressor genes. Conversely, hypermethylation of H3K27 occurs in late-stage epithelial ovarian cancer, which promotes tumor vascularization and tumor cell migration. By using each cancer type as a case study, this review emphasizes the importance of H3K27me3 in cancer while demonstrating that the mechanisms of histone H3 modification and subsequent gene expression changes are not a one-size-fits-all across cancer types.
Collapse
Affiliation(s)
- Charles A. Day
- Hormel Institute, University of Minnesota, Austin, MN 55912, USA
- Mayo Clinic, Rochester, MN 55902, USA
- Correspondence:
| | - Edward H. Hinchcliffe
- Hormel Institute, University of Minnesota, Austin, MN 55912, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - James P. Robinson
- Hormel Institute, University of Minnesota, Austin, MN 55912, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
8
|
Schwark K, Messinger D, Cummings JR, Bradin J, Kawakibi A, Babila CM, Lyons S, Ji S, Cartaxo RT, Kong S, Cantor E, Koschmann C, Yadav VN. Receptor tyrosine kinase (RTK) targeting in pediatric high-grade glioma and diffuse midline glioma: Pre-clinical models and precision medicine. Front Oncol 2022; 12:922928. [PMID: 35978801 PMCID: PMC9376238 DOI: 10.3389/fonc.2022.922928] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Pediatric high-grade glioma (pHGG), including both diffuse midline glioma (DMG) and non-midline tumors, continues to be one of the deadliest oncologic diagnoses (both henceforth referred to as “pHGG”). Targeted therapy options aimed at key oncogenic receptor tyrosine kinase (RTK) drivers using small-molecule RTK inhibitors has been extensively studied, but the absence of proper in vivo modeling that recapitulate pHGG biology has historically been a research challenge. Thankfully, there have been many recent advances in animal modeling, including Cre-inducible transgenic models, as well as intra-uterine electroporation (IUE) models, which closely recapitulate the salient features of human pHGG tumors. Over 20% of pHGG have been found in sequencing studies to have alterations in platelet derived growth factor-alpha (PDGFRA), making growth factor modeling and inhibition via targeted tyrosine kinases a rich vein of interest. With commonly found alterations in other growth factors, including FGFR, EGFR, VEGFR as well as RET, MET, and ALK, it is necessary to model those receptors, as well. Here we review the recent advances in murine modeling and precision targeting of the most important RTKs in their clinical context. We additionally provide a review of current work in the field with several small molecule RTK inhibitors used in pre-clinical or clinical settings for treatment of pHGG.
Collapse
Affiliation(s)
- Kallen Schwark
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Dana Messinger
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Jessica R. Cummings
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Joshua Bradin
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Abed Kawakibi
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Clarissa M. Babila
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Samantha Lyons
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Sunjong Ji
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Rodrigo T. Cartaxo
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Seongbae Kong
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Evan Cantor
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Carl Koschmann
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Viveka Nand Yadav
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI, United States
- Department of Pediatrics, Children's Mercy Research Institute (CMRI), Kansas, MO, United States
- Department of Pediatrics, University of Missouri Kansas City School of Medicine, Kansas, MO, United States
- *Correspondence: Viveka Nand Yadav,
| |
Collapse
|
9
|
Dhar S, Gadd S, Patel P, Vaynshteyn J, Raju GP, Hashizume R, Brat DJ, Becher OJ. A tumor suppressor role for EZH2 in diffuse midline glioma pathogenesis. Acta Neuropathol Commun 2022; 10:47. [PMID: 35395831 PMCID: PMC8994223 DOI: 10.1186/s40478-022-01336-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/21/2022] [Indexed: 11/10/2022] Open
Abstract
Pediatric high-grade gliomas, specifically diffuse midline gliomas, account for only 20% of clinical cases but are 100% fatal. A majority of the DMG cases are characterized by the signature K27M mutation in histone H3. The H3K27M mutation opposes the function of enhancer of zeste homolog 2 (EZH2), the methyltransferase enzyme of the polycomb repressor complex 2. However, the role of EZH2 in DMG pathogenesis is unclear. In this study, we demonstrate a tumor suppressor function for EZH2 using Ezh2 loss- and gain-of-function studies in H3WT DMG mouse models. Genetic ablation of Ezh2 increased cell proliferation and tumor grade while expression of an Ezh2 gain-of-function mutation significantly reduced tumor incidence and increased tumor latency. Transcriptomic analysis revealed that Ezh2 deletion upregulates an inflammatory response with upregulation of immunoproteasome genes such as Psmb8, Psmb9, and Psmb10. Ezh2 gain-of-function resulted in enrichment of the oxidative phosphorylation/mitochondrial metabolic pathway namely the isocitrate dehydrogenase Idh1/2/3 genes. Pharmacological inhibition of EZH2 augmented neural progenitor cell proliferation, supporting the tumor suppressive role of EZH2. In vivo 7-day treatment of H3K27M DMG tumor bearing mice with an EZH2 inhibitor, Tazemetostat, did not alter proliferation or significantly impact survival. Together our results suggest that EZH2 has a tumor suppressor function in DMG and warrants caution in clinical translation of EZH2 inhibitors to treat patients with DMG.
Collapse
Affiliation(s)
- Swati Dhar
- grid.413808.60000 0004 0388 2248Department of Pediatrics, Simpson Querrey Biomedical Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital, Chicago, IL USA ,Present Address: NeoImmuneTech Inc., 2400 Research Blvd., Suite 250, Rockville, MD USA
| | - Samantha Gadd
- grid.413808.60000 0004 0388 2248Department of Pathology, Simpson Querrey Biomedical Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital, Chicago, IL USA
| | - Priyam Patel
- grid.16753.360000 0001 2299 3507Quantitative Data Science Core, Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Jake Vaynshteyn
- grid.59734.3c0000 0001 0670 2351Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - G. Praveen Raju
- grid.59734.3c0000 0001 0670 2351Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Rintaro Hashizume
- grid.413808.60000 0004 0388 2248Department of Pediatrics, Simpson Querrey Biomedical Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital, Chicago, IL USA ,grid.16753.360000 0001 2299 3507Department of Pediatrics and Department of Biochemistry and Molecular Biology, Simpson Querrey Biomedical Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital, Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Daniel J. Brat
- grid.16753.360000 0001 2299 3507Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Oren J. Becher
- grid.413808.60000 0004 0388 2248Department of Pediatrics, Simpson Querrey Biomedical Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital, Chicago, IL USA ,grid.16753.360000 0001 2299 3507Department of Pediatrics and Department of Biochemistry and Molecular Biology, Simpson Querrey Biomedical Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital, Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL USA ,grid.59734.3c0000 0001 0670 2351Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| |
Collapse
|
10
|
Khadka P, Reitman ZJ, Lu S, Buchan G, Gionet G, Dubois F, Carvalho DM, Shih J, Zhang S, Greenwald NF, Zack T, Shapira O, Pelton K, Hartley R, Bear H, Georgis Y, Jarmale S, Melanson R, Bonanno K, Schoolcraft K, Miller PG, Condurat AL, Gonzalez EM, Qian K, Morin E, Langhnoja J, Lupien LE, Rendo V, Digiacomo J, Wang D, Zhou K, Kumbhani R, Guerra Garcia ME, Sinai CE, Becker S, Schneider R, Vogelzang J, Krug K, Goodale A, Abid T, Kalani Z, Piccioni F, Beroukhim R, Persky NS, Root DE, Carcaboso AM, Ebert BL, Fuller C, Babur O, Kieran MW, Jones C, Keshishian H, Ligon KL, Carr SA, Phoenix TN, Bandopadhayay P. PPM1D mutations are oncogenic drivers of de novo diffuse midline glioma formation. Nat Commun 2022; 13:604. [PMID: 35105861 PMCID: PMC8807747 DOI: 10.1038/s41467-022-28198-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/07/2022] [Indexed: 12/23/2022] Open
Abstract
The role of PPM1D mutations in de novo gliomagenesis has not been systematically explored. Here we analyze whole genome sequences of 170 pediatric high-grade gliomas and find that truncating mutations in PPM1D that increase the stability of its phosphatase are clonal driver events in 11% of Diffuse Midline Gliomas (DMGs) and are enriched in primary pontine tumors. Through the development of DMG mouse models, we show that PPM1D mutations potentiate gliomagenesis and that PPM1D phosphatase activity is required for in vivo oncogenesis. Finally, we apply integrative phosphoproteomic and functional genomics assays and find that oncogenic effects of PPM1D truncation converge on regulators of cell cycle, DNA damage response, and p53 pathways, revealing therapeutic vulnerabilities including MDM2 inhibition.
Collapse
Affiliation(s)
- Prasidda Khadka
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA, 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Biological and Biomedical Sciences PhD Program, Harvard University, Cambridge, MA, 02138, USA
| | - Zachary J Reitman
- Department of Radiation Oncology, Duke University, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University, Durham, NC, 27710, USA
- The Preston Robert Tisch Brain Tumor Center at Duke, Duke University, Durham, NC, 27710, USA
| | - Sophie Lu
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02215, USA
| | - Graham Buchan
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02215, USA
| | - Gabrielle Gionet
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02215, USA
| | - Frank Dubois
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA, 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Diana M Carvalho
- Division of Molecular Pathology, Institute of Cancer Research, London, UK
| | - Juliann Shih
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Shu Zhang
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Noah F Greenwald
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA, 02215, USA
| | - Travis Zack
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA, 02215, USA
| | - Ofer Shapira
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA, 02215, USA
| | - Kristine Pelton
- Department of Oncologic Pathology, Dana Farber Cancer Institute, Boston, MA, 02215, USA
| | - Rachel Hartley
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Heather Bear
- Research in Patient Services, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45267, USA
| | - Yohanna Georgis
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02215, USA
| | - Spandana Jarmale
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02215, USA
| | - Randy Melanson
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Kevin Bonanno
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Kathleen Schoolcraft
- Department of Oncologic Pathology, Dana Farber Cancer Institute, Boston, MA, 02215, USA
| | - Peter G Miller
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Alexandra L Condurat
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02215, USA
| | - Elizabeth M Gonzalez
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02215, USA
| | - Kenin Qian
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02215, USA
| | - Eric Morin
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02215, USA
| | - Jaldeep Langhnoja
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Leslie E Lupien
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02215, USA
| | - Veronica Rendo
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA, 02215, USA
| | - Jeromy Digiacomo
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02215, USA
| | - Dayle Wang
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02215, USA
| | - Kevin Zhou
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02215, USA
| | - Rushil Kumbhani
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02215, USA
| | | | - Claire E Sinai
- Department of Oncologic Pathology, Dana Farber Cancer Institute, Boston, MA, 02215, USA
| | - Sarah Becker
- Department of Oncologic Pathology, Dana Farber Cancer Institute, Boston, MA, 02215, USA
| | - Rachel Schneider
- Department of Oncologic Pathology, Dana Farber Cancer Institute, Boston, MA, 02215, USA
| | - Jayne Vogelzang
- Department of Oncologic Pathology, Dana Farber Cancer Institute, Boston, MA, 02215, USA
| | - Karsten Krug
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Amy Goodale
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Tanaz Abid
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Zohra Kalani
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | | | - Rameen Beroukhim
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA, 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Nicole S Persky
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - David E Root
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Angel M Carcaboso
- Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Institut de Recerca Sant Joan de Deu, Barcelona, 08950, Spain
| | - Benjamin L Ebert
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA
| | - Christine Fuller
- Department of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45267, USA
| | - Ozgun Babur
- College of Science and Mathematics, University of Massachusetts Boston, Boston, MA, 02125, USA
| | - Mark W Kieran
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02215, USA
- Bristol Myers Squibb, Boston, Devens, MA, 01434, USA
| | - Chris Jones
- Division of Molecular Pathology, Institute of Cancer Research, London, UK
| | | | - Keith L Ligon
- Department of Oncologic Pathology, Dana Farber Cancer Institute, Boston, MA, 02215, USA
| | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Timothy N Phoenix
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, 45267, USA.
- Research in Patient Services, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45267, USA.
| | - Pratiti Bandopadhayay
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02215, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
11
|
Haydar D, Ibañez-Vega J, Krenciute G. T-Cell Immunotherapy for Pediatric High-Grade Gliomas: New Insights to Overcoming Therapeutic Challenges. Front Oncol 2021; 11:718030. [PMID: 34760690 PMCID: PMC8573171 DOI: 10.3389/fonc.2021.718030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 10/08/2021] [Indexed: 01/06/2023] Open
Abstract
Despite decades of research, pediatric central nervous system (CNS) tumors remain the most debilitating, difficult to treat, and deadliest cancers. Current therapies, including radiation, chemotherapy, and/or surgery, are unable to cure these diseases and are associated with serious adverse effects and long-term impairments. Immunotherapy using chimeric antigen receptor (CAR) T cells has the potential to elucidate therapeutic antitumor immune responses that improve survival without the devastating adverse effects associated with other therapies. Yet, despite the outstanding performance of CAR T cells against hematologic malignancies, they have shown little success targeting brain tumors. This lack of efficacy is due to a scarcity of targetable antigens, interactions with the immune microenvironment, and physical and biological barriers limiting the homing and trafficking of CAR T cells to brain tumors. In this review, we summarize experiences with CAR T-cell therapy for pediatric CNS tumors in preclinical and clinical settings and focus on the current roadblocks and novel strategies to potentially overcome those therapeutic challenges.
Collapse
Affiliation(s)
| | | | - Giedre Krenciute
- Department of Bone Marrow Transplantation & Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN, United States
| |
Collapse
|
12
|
Wei X, Meel MH, Breur M, Bugiani M, Hulleman E, Phoenix TN. Defining tumor-associated vascular heterogeneity in pediatric high-grade and diffuse midline gliomas. Acta Neuropathol Commun 2021; 9:142. [PMID: 34425907 PMCID: PMC8381557 DOI: 10.1186/s40478-021-01243-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/10/2021] [Indexed: 12/23/2022] Open
Abstract
The blood–brain barrier (BBB) plays important roles in brain tumor pathogenesis and treatment response, yet our understanding of its function and heterogeneity within or across brain tumor types remains poorly characterized. Here we analyze the neurovascular unit (NVU) of pediatric high-grade glioma (pHGG) and diffuse midline glioma (DMG) using patient derived xenografts and natively forming glioma mouse models. We show tumor-associated vascular differences between these glioma subtypes, and parallels between PDX and mouse model systems, with DMG models maintaining a more normal vascular architecture, BBB function and endothelial transcriptional program relative to pHGG models. Unlike prior work in angiogenic brain tumors, we find that expression of secreted Wnt antagonists do not alter the tumor-associated vascular phenotype in DMG tumor models. Together, these findings highlight vascular heterogeneity between pHGG and DMG and differences in their response to alterations in developmental BBB signals that may participate in driving these pathological differences.
Collapse
|
13
|
Wei X, Jessa S, Kleinman CL, Phoenix TN. Mapping Angiopoietin1 expression in the developing and adult brain. Dev Neurosci 2021; 43:321-334. [PMID: 34348288 DOI: 10.1159/000518351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 07/06/2021] [Indexed: 11/19/2022] Open
Affiliation(s)
- Xin Wei
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio, USA,
| | - Selin Jessa
- Quantitative Life Sciences, McGill University, Montreal, Québec, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Québec, Canada
| | - Claudia L Kleinman
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Québec, Canada
- Department of Human Genetics, McGill University, Montreal, Québec, Canada
| | - Timothy N Phoenix
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio, USA
- Research in Patient Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
14
|
Garcia-Fabiani MB, Haase S, Comba A, Carney S, McClellan B, Banerjee K, Alghamri MS, Syed F, Kadiyala P, Nunez FJ, Candolfi M, Asad A, Gonzalez N, Aikins ME, Schwendeman A, Moon JJ, Lowenstein PR, Castro MG. Genetic Alterations in Gliomas Remodel the Tumor Immune Microenvironment and Impact Immune-Mediated Therapies. Front Oncol 2021; 11:631037. [PMID: 34168976 PMCID: PMC8217836 DOI: 10.3389/fonc.2021.631037] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 05/06/2021] [Indexed: 12/13/2022] Open
Abstract
High grade gliomas are malignant brain tumors that arise in the central nervous system, in patients of all ages. Currently, the standard of care, entailing surgery and chemo radiation, exhibits a survival rate of 14-17 months. Thus, there is an urgent need to develop new therapeutic strategies for these malignant brain tumors. Currently, immunotherapies represent an appealing approach to treat malignant gliomas, as the pre-clinical data has been encouraging. However, the translation of the discoveries from the bench to the bedside has not been as successful as with other types of cancer, and no long-lasting clinical benefits have been observed for glioma patients treated with immune-mediated therapies so far. This review aims to discuss our current knowledge about gliomas, their molecular particularities and the impact on the tumor immune microenvironment. Also, we discuss several murine models used to study these therapies pre-clinically and how the model selection can impact the outcomes of the approaches to be tested. Finally, we present different immunotherapy strategies being employed in clinical trials for glioma and the newest developments intended to harness the immune system against these incurable brain tumors.
Collapse
Affiliation(s)
- Maria B. Garcia-Fabiani
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Santiago Haase
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Andrea Comba
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Stephen Carney
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Brandon McClellan
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Immunology graduate program, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Kaushik Banerjee
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Mahmoud S. Alghamri
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Faisal Syed
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Padma Kadiyala
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | | | - Marianela Candolfi
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Antonela Asad
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nazareno Gonzalez
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marisa E. Aikins
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
| | - James J. Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Pedro R. Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Maria G. Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
15
|
Metselaar DS, du Chatinier A, Stuiver I, Kaspers GJL, Hulleman E. Radiosensitization in Pediatric High-Grade Glioma: Targets, Resistance and Developments. Front Oncol 2021; 11:662209. [PMID: 33869066 PMCID: PMC8047603 DOI: 10.3389/fonc.2021.662209] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/17/2021] [Indexed: 12/25/2022] Open
Abstract
Pediatric high-grade gliomas (pHGG) are the leading cause of cancer-related death in children. These epigenetically dysregulated tumors often harbor mutations in genes encoding histone 3, which contributes to a stem cell-like, therapy-resistant phenotype. Furthermore, pHGG are characterized by a diffuse growth pattern, which, together with their delicate location, makes complete surgical resection often impossible. Radiation therapy (RT) is part of the standard therapy against pHGG and generally the only modality, apart from surgery, to provide symptom relief and a delay in tumor progression. However, as a single treatment modality, RT still offers no chance for a cure. As with most therapeutic approaches, irradiated cancer cells often acquire resistance mechanisms that permit survival or stimulate regrowth after treatment, thereby limiting the efficacy of RT. Various preclinical studies have investigated radiosensitizers in pHGG models, without leading to an improved clinical outcome for these patients. However, our recently improved molecular understanding of pHGG generates new opportunities to (re-)evaluate radiosensitizers in these malignancies. Furthermore, the use of radio-enhancing agents has several benefits in pHGG compared to other cancers, which will be discussed here. This review provides an overview and a critical evaluation of the radiosensitization strategies that have been studied to date in pHGG, thereby providing a framework for improving radiosensitivity of these rapidly fatal brain tumors.
Collapse
Affiliation(s)
- Dennis S Metselaar
- Department of Neuro-oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Aimée du Chatinier
- Department of Neuro-oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Iris Stuiver
- Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Gertjan J L Kaspers
- Department of Neuro-oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Esther Hulleman
- Department of Neuro-oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| |
Collapse
|
16
|
Biery MC, Noll A, Myers C, Morris SM, Winter CA, Pakiam F, Cole BL, Browd SR, Olson JM, Vitanza NA. A Protocol for the Generation of Treatment-naïve Biopsy-derived Diffuse Intrinsic Pontine Glioma and Diffuse Midline Glioma Models. JOURNAL OF EXPERIMENTAL NEUROLOGY 2020. [PMID: 33768215 PMCID: PMC7990285 DOI: 10.33696//neurol.1.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a universally fatal tumor of the brainstem, most commonly affecting young children. Due to its location, surgical resection is not achievable, but consideration of a biopsy has become standard practice at children’s hospitals with the appropriate neurosurgical expertise. While the decision to obtain a biopsy should be directed by the presence of atypical radiographic features that call the diagnosis of DIPG into question or the requirement of biopsy tissue for clinical trial enrollment, once this precious tissue is available its use for research should be considered. The majority of DIPG and diffuse midline glioma, H3 K27M-mutant (DMG) models are autopsy-derived or genetically-engineered, each of which has limitations for translational studies, so the use of biopsy tissue for laboratory model development provides an opportunity to create unique model systems. Here, we present a detailed laboratory protocol for the generation of treatment-naïve biopsy-derived DIPG/DMG models.
Collapse
Affiliation(s)
- Matt C Biery
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Alyssa Noll
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Molecular and Cellular Biology Graduate Program and Medical Scientist Training Program, University of Washington, Seattle, WA, USA
| | - Carrie Myers
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Conrad A Winter
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Department of Laboratories, Seattle Children's Hospital, Seattle, WA, USA
| | - Fiona Pakiam
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Bonnie L Cole
- Department of Laboratories, Seattle Children's Hospital, Seattle, WA, USA.,Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Samuel R Browd
- Division of Neurosurgery, Department of Neurological Surgery, University of Washington, Seattle Children's Hospital, Seattle, WA, USA
| | - James M Olson
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Division of Hematology/Oncology, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | - Nicholas A Vitanza
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Division of Hematology/Oncology, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| |
Collapse
|
17
|
Faisal SM, Mendez FM, Nunez F, Castro MG, Lowenstein PR. Immune-stimulatory (TK/Flt3L) gene therapy opens the door to a promising new treatment strategy against brainstem gliomas. Oncotarget 2020; 11:4607-4612. [PMID: 33400737 PMCID: PMC7747859 DOI: 10.18632/oncotarget.27834] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 11/18/2020] [Indexed: 11/25/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a rare brainstem tumor which carries a dismal prognosis. To date. there are no effective treatments for DIPG. Transcriptomic studies have shown that DIPGs have a distinct profile compared to hemispheric high-grade pediatric gliomas. These specific genomic features coupled with the younger median age group suggest that DIPG is of developmental origin. There is a major unmet need for novel effective therapeutic approaches for DIPG. Clinical and preclinical studies have expanded our understanding of the molecular pathways in this deadly disease. We have developed a genetically engineered brainstem glioma model harboring the recurrent DIPG mutation, activin A receptor type I (ACVR1)-G328V (mACVR1) using the sleeping beauty transposon system. DIPG neurospheres isolated from the genetically engineered mouse model were implanted into the pons of immune-competent mice to assess the therapeutic efficacy and toxicity of immunostimulatory gene therapy using adenoviruses expressing thymidine kinase (TK) and fms-like tyrosine kinase 3 ligand (Flt3L). Immunostimulatory adenoviral-mediated delivery of TK/Flt3L in mice bearing brainstem gliomas resulted in antitumor immunity, recruitment of antitumor-specific T cells, and improved median survival by stimulating the host antitumor immune response. Therapeutic efficacy of the immunostimulatory gene therapy strategy will be tested in the clinical arena in a Phase I clinical trial. We also discuss immunotherapeutic interventions currently being implemented in DIPG patients and discuss the profound therapeutic implications of immunotherapy for this patient populations.
Collapse
Affiliation(s)
- Syed M. Faisal
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Flor M. Mendez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Fernando Nunez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maria G. Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Pedro R. Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
18
|
Epigenetic activation of a RAS/MYC axis in H3.3K27M-driven cancer. Nat Commun 2020; 11:6216. [PMID: 33277484 PMCID: PMC7718276 DOI: 10.1038/s41467-020-19972-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 11/10/2020] [Indexed: 12/20/2022] Open
Abstract
Histone H3 lysine 27 (H3K27M) mutations represent the canonical oncohistone, occurring frequently in midline gliomas but also identified in haematopoietic malignancies and carcinomas. H3K27M functions, at least in part, through widespread changes in H3K27 trimethylation but its role in tumour initiation remains obscure. To address this, we created a transgenic mouse expressing H3.3K27M in diverse progenitor cell populations. H3.3K27M expression drives tumorigenesis in multiple tissues, which is further enhanced by Trp53 deletion. We find that H3.3K27M epigenetically activates a transcriptome, enriched for PRC2 and SOX10 targets, that overrides developmental and tissue specificity and is conserved between H3.3K27M-mutant mouse and human tumours. A key feature of the H3K27M transcriptome is activation of a RAS/MYC axis, which we find can be targeted therapeutically in isogenic and primary DIPG cell lines with H3.3K27M mutations, providing an explanation for the common co-occurrence of alterations in these pathways in human H3.3K27M-driven cancer. Taken together, these results show how H3.3K27M-driven transcriptome remodelling promotes tumorigenesis and will be critical for targeting cancers with these mutations. Histone H3 at lysine 27 (H3K27M) is often mutated in cancer but its role in tumour initiation is unclear. Here, the authors generated a transgenic model expressing H3.3K27M from the Fabp7 gene promoter, demonstrating that H3.3K27M can initiate diverse tumorigesis on its own, acting through a RAS/MYC transcriptomic programme.
Collapse
|
19
|
Biery MC, Noll A, Myers C, Morris SM, Winter CA, Pakiam F, Cole BL, Browd SR, Olson JM, Vitanza NA. A Protocol for the Generation of Treatment-naïve Biopsy-derived Diffuse Intrinsic Pontine Glioma and Diffuse Midline Glioma Models. JOURNAL OF EXPERIMENTAL NEUROLOGY 2020; 1:158-167. [PMID: 33768215 PMCID: PMC7990285 DOI: 10.33696/neurol.1.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a universally fatal tumor of the brainstem, most commonly affecting young children. Due to its location, surgical resection is not achievable, but consideration of a biopsy has become standard practice at children's hospitals with the appropriate neurosurgical expertise. While the decision to obtain a biopsy should be directed by the presence of atypical radiographic features that call the diagnosis of DIPG into question or the requirement of biopsy tissue for clinical trial enrollment, once this precious tissue is available its use for research should be considered. The majority of DIPG and diffuse midline glioma, H3 K27M-mutant (DMG) models are autopsy-derived or genetically-engineered, each of which has limitations for translational studies, so the use of biopsy tissue for laboratory model development provides an opportunity to create unique model systems. Here, we present a detailed laboratory protocol for the generation of treatment-naïve biopsy-derived DIPG/DMG models.
Collapse
Affiliation(s)
- Matt C. Biery
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Alyssa Noll
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Molecular and Cellular Biology Graduate Program and Medical Scientist Training Program, University of Washington, Seattle, WA, USA
| | - Carrie Myers
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Conrad A. Winter
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Laboratories, Seattle Children’s Hospital, Seattle, WA, USA
| | - Fiona Pakiam
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Bonnie L. Cole
- Department of Laboratories, Seattle Children’s Hospital, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Samuel R. Browd
- Division of Neurosurgery, Department of Neurological Surgery, University of Washington, Seattle Children’s Hospital, Seattle, WA, USA
| | - James M. Olson
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Seattle Children’s Hospital, University of Washington, Seattle, WA, USA
| | - Nicholas A. Vitanza
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Seattle Children’s Hospital, University of Washington, Seattle, WA, USA
| |
Collapse
|
20
|
Miklja Z, Yadav VN, Cartaxo RT, Siada R, Thomas CC, Cummings JR, Mullan B, Stallard S, Paul A, Bruzek AK, Wierzbicki K, Yang T, Garcia T, Wolfe I, Leonard M, Robertson PL, Garton HJ, Wahl DR, Parmar H, Sarkaria JN, Kline C, Mueller S, Nicolaides T, Glasser C, Leary SE, Venneti S, Kumar-Sinha C, Chinnaiyan AM, Mody R, Pai MP, Phoenix TN, Marini BL, Koschmann C. Everolimus improves the efficacy of dasatinib in PDGFRα-driven glioma. J Clin Invest 2020; 130:5313-5325. [PMID: 32603316 PMCID: PMC7524471 DOI: 10.1172/jci133310] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 06/24/2020] [Indexed: 12/26/2022] Open
Abstract
Pediatric and adult high-grade gliomas (HGGs) frequently harbor PDGFRA alterations. We hypothesized that cotreatment with everolimus may improve the efficacy of dasatinib in PDGFRα-driven glioma through combinatorial synergism and increased tumor accumulation of dasatinib. We performed dose-response, synergism, P-glycoprotein inhibition, and pharmacokinetic studies in in vitro and in vivo human and mouse models of HGG. Six patients with recurrent PDGFRα-driven glioma were treated with dasatinib and everolimus. We found that dasatinib effectively inhibited the proliferation of mouse and human primary HGG cells with a variety of PDGFRA alterations. Dasatinib exhibited synergy with everolimus in the treatment of HGG cells at low nanomolar concentrations of both agents, with a reduction in mTOR signaling that persisted after dasatinib treatment alone. Prolonged exposure to everolimus significantly improved the CNS retention of dasatinib and extended the survival of PPK tumor-bearing mice (mutant TP53, mutant PDGFRA, H3K27M). Six pediatric patients with glioma tolerated this combination without significant adverse events, and 4 patients with recurrent disease (n = 4) had a median overall survival of 8.5 months. Our results show that the efficacy of dasatinib treatment of PDGFRα-driven HGG was enhanced with everolimus and suggest a promising route for improving targeted therapy for this patient population.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Hemant Parmar
- Department of Radiology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Jann N. Sarkaria
- Department of Oncology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Cassie Kline
- Department of Pediatrics and
- Department of Neurology, UCSF, San Francisco, California, USA
| | - Sabine Mueller
- Department of Pediatrics and
- Department of Neurology, UCSF, San Francisco, California, USA
| | - Theodore Nicolaides
- Division of Pediatric Hematology/Oncology, NYU Langone Medical Center, New York, New York, USA
| | - Chana Glasser
- Division of Pediatric Hematology/Oncology, NYU Winthrop Hospital, Mineola, New York, USA
| | - Sarah E.S. Leary
- Seattle Children’s Hospital/University of Washington (UW), Seattle, Washington, USA
| | | | | | - Arul M. Chinnaiyan
- Department of Pathology
- Department of Urology
- Michigan Center for Translational Pathology
- Howard Hughes Medical Institute
- Rogel Cancer Center, and
| | | | - Manjunath P. Pai
- College of Pharmacy, Michigan Medicine, Ann Arbor, Michigan, USA
| | | | | | | |
Collapse
|
21
|
Graham MS, Mellinghoff IK. Histone-Mutant Glioma: Molecular Mechanisms, Preclinical Models, and Implications for Therapy. Int J Mol Sci 2020; 21:E7193. [PMID: 33003625 PMCID: PMC7582376 DOI: 10.3390/ijms21197193] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/21/2020] [Accepted: 09/25/2020] [Indexed: 02/06/2023] Open
Abstract
Pediatric high-grade glioma (pHGG) is the leading cause of cancer death in children. Despite histologic similarities, it has recently become apparent that this disease is molecularly distinct from its adult counterpart. Specific hallmark oncogenic histone mutations within pediatric malignant gliomas divide these tumors into subgroups with different neuroanatomic and chronologic predilections. In this review, we will summarize the characteristic molecular alterations of pediatric high-grade gliomas, with a focus on how preclinical models of these alterations have furthered our understanding of their oncogenicity as well as their potential impact on developing targeted therapies for this devastating disease.
Collapse
Affiliation(s)
- Maya S. Graham
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Ingo K. Mellinghoff
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
22
|
Chung C, Sweha SR, Pratt D, Tamrazi B, Panwalkar P, Banda A, Bayliss J, Hawes D, Yang F, Lee HJ, Shan M, Cieslik M, Qin T, Werner CK, Wahl DR, Lyssiotis CA, Bian Z, Shotwell JB, Yadav VN, Koschmann C, Chinnaiyan AM, Blüml S, Judkins AR, Venneti S. Integrated Metabolic and Epigenomic Reprograming by H3K27M Mutations in Diffuse Intrinsic Pontine Gliomas. Cancer Cell 2020; 38:334-349.e9. [PMID: 32795401 PMCID: PMC7494613 DOI: 10.1016/j.ccell.2020.07.008] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 05/28/2020] [Accepted: 07/15/2020] [Indexed: 01/08/2023]
Abstract
H3K27M diffuse intrinsic pontine gliomas (DIPGs) are fatal and lack treatments. They mainly harbor H3.3K27M mutations resulting in H3K27me3 reduction. Integrated analysis in H3.3K27M cells, tumors, and in vivo imaging in patients showed enhanced glycolysis, glutaminolysis, and tricarboxylic acid cycle metabolism with high alpha-ketoglutarate (α-KG) production. Glucose and/or glutamine-derived α-KG maintained low H3K27me3 in H3.3K27M cells, and inhibition of key enzymes in glycolysis or glutaminolysis increased H3K27me3, altered chromatin accessibility, and prolonged survival in animal models. Previous studies have shown that mutant isocitrate-dehydrogenase (mIDH)1/2 glioma cells convert α-KG to D-2-hydroxyglutarate (D-2HG) to increase H3K27me3. Here, we show that H3K27M and IDH1 mutations are mutually exclusive and experimentally synthetic lethal. Overall, we demonstrate that H3.3K27M and mIDH1 hijack a conserved and critical metabolic pathway in opposing ways to maintain their preferred epigenetic state. Consequently, interruption of this metabolic/epigenetic pathway showed potent efficacy in preclinical models, suggesting key therapeutic targets for much needed treatments.
Collapse
Affiliation(s)
- Chan Chung
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Michigan Medicine, University of Michigan, 3520E MSRB 1, 1150 West Medical Center Drive, Ann Arbor, MI 48109-41804, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Stefan R Sweha
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Michigan Medicine, University of Michigan, 3520E MSRB 1, 1150 West Medical Center Drive, Ann Arbor, MI 48109-41804, USA; Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Drew Pratt
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Michigan Medicine, University of Michigan, 3520E MSRB 1, 1150 West Medical Center Drive, Ann Arbor, MI 48109-41804, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Benita Tamrazi
- Department of Radiology, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| | - Pooja Panwalkar
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Michigan Medicine, University of Michigan, 3520E MSRB 1, 1150 West Medical Center Drive, Ann Arbor, MI 48109-41804, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Adam Banda
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Michigan Medicine, University of Michigan, 3520E MSRB 1, 1150 West Medical Center Drive, Ann Arbor, MI 48109-41804, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jill Bayliss
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Michigan Medicine, University of Michigan, 3520E MSRB 1, 1150 West Medical Center Drive, Ann Arbor, MI 48109-41804, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Debra Hawes
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Fusheng Yang
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ho-Joon Lee
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Mengrou Shan
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Marcin Cieslik
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Michigan Center for Translational Pathology, Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Tingting Qin
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Christian K Werner
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Daniel R Wahl
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Zhiguo Bian
- Centralized Medicinal Chemistry, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - J Brad Shotwell
- Centralized Medicinal Chemistry, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - Viveka Nand Yadav
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Carl Koschmann
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Michigan Center for Translational Pathology, Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Stefan Blüml
- Department of Radiology, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| | - Alexander R Judkins
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Sriram Venneti
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Michigan Medicine, University of Michigan, 3520E MSRB 1, 1150 West Medical Center Drive, Ann Arbor, MI 48109-41804, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
23
|
Affiliation(s)
- Robert Siddaway
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.,Division of Pathology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Cynthia Hawkins
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.,Division of Pathology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|