Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
Oncology Reports
Join Editorial Board Propose a Special Issue
Print ISSN: 1021-335X Online ISSN: 1791-2431
Journal Cover
March-2026 Volume 55 Issue 3

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
March-2026 Volume 55 Issue 3

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Epigenetic mechanisms and therapeutic advances in diffuse midline glioma (Review)

  • Authors:
    • Wenbo Wu
    • Wenlin Chen
    • Wenbin Ma
    • Yu Wang
  • View Affiliations / Copyright

    Affiliations: Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital (East), Beijing 100730, P.R. China
    Copyright: © Wu et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].
  • Article Number: 50
    |
    Published online on: January 20, 2026
       https://doi.org/10.3892/or.2026.9055
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:


Abstract

Gliomas are the most common primary malignant tumors of the central nervous system in adults, with diffuse midline gliomas (DMG) being particularly aggressive and associated with inferior survival rate. Notwithstanding advances in molecular diagnostics and epigenetics, the specific pathological mechanisms of DMG remain to be fully elucidated. A series of studies have demonstrated that histone modifications, particularly the histone H3 lysine 27 (H3K27)M mutation, play a pivotal role in the development and progression of DMG. The mutation disrupts histone methylation and acetylation to induce widespread gene expression abnormalities, tumor aggressiveness and treatment resistance. Conventional treatments such as surgery, local radiotherapy and chemotherapy offer limited efficacy. However, emerging precision therapies targeting histone mutations, epigenetic modifications and innovative immunotherapies show promise in improving outcomes. The present study provided a comprehensive overview of the molecular mechanisms, epigenetic characteristics and the latest therapeutic advances in DMG. By investigating the H3K27M mutation and its associated epigenetic mechanisms, the present review aimed to establish theoretical frameworks and research avenues for developing precise therapeutic strategies for DMG, thus contributing to advancing the field of personalized medicine.

Introduction

Gliomas are the most prevalent primary malignant tumors of the central nervous system (CNS) in adults (1), with diffuse midline glioma (DMG) standing out for its high invasiveness and abysmal prognosis. Driven by the histone H3 lysine 27 (H3K27)M mutation, DMG exhibits aggressive behavior and marked resistance to treatment, making it a formidable challenge in neuro-oncology.

DMG predominantly affects children and adolescents, whereas adult cases are uncommon. Clinically, numerous pediatric DMGs present as diffuse intrinsic pontine glioma (DIPG), a radiographic entity centered in the pons. Epidemiologic estimates suggest 20–30 new DIPG cases annually in the UK (2) and 100–400 annually in the US (2,3) and a Canadian population-based cohort identified 143 pediatric DIPG cases over a 10-year period (~14 cases per year) (2). DMG remains highly lethal, with a median overall survival (OS) of ~11 months (4) and 1-, 2- and 5-year survival rates of 42.3, 9.6 and 2.2% in a 1,008-case registry cohort (5).

DMGs are invasive tumors affecting midline structures such as the thalamus, pons, brainstem and spinal cord. Clinical manifestations vary by location, including cranial nerve dysfunction, ataxia and raised intracranial pressure. MRI is key for diagnosis, showing heterogeneous features such as diffuse or distinct boundaries, variable contrast enhancement and similarities to low-grade astrocytomas (6). Pons-involved DMGs often infiltrate >50% of the pons and may encase the basilar artery, with minimal T1 contrast enhancement and uneven T1/T2 signal intensities, often with necrosis or hemorrhage (7,8), reflecting their histopathological heterogeneity.

Molecular pathological studies of DMG have revealed its unique epigenetic characteristics, focusing on the pivotal role of the H3K27M mutation in its pathogenesis (9). This mutation replaces lysine 27 of histone H3 with methionine, substantially reducing H3K27 trimethylation (H3K27me3) and causing widespread gene expression reprogramming. It disrupts epigenetic regulation, promoting tumor proliferation, invasion and treatment resistance (10). Additionally, the H3K27M mutation dysregulates multiple key signaling pathways, including MAPK and PI3K/AKT, driving rapid growth and creating a tumor-supportive micro-environment (11). These findings establish H3K27M mutation as a central driver of DMG and underscore the urgent need for targeted therapeutic strategies.

Despite notable advances in understanding the molecular basis of DMG, the therapeutic landscape for this disease remains beset with formidable challenges. Surgery, radiotherapy and chemotherapy provide minimal improvement in overall survival currently, as epigenetic alterations driven by the H3K27M mutation render these tumors highly resistant to conventional treatments. However, emerging targeted therapies and immunotherapies, including CAR-T cell therapy, have garnered considerable research interest (12) though they require further clinical validation.

Against this backdrop, the present review systematically summarized the molecular characteristics, genetic signatures and latest progress in DMG treatment, emphasizing the effect of histone alterations and innovative therapeutic strategies to deepen the understanding of DMG and provide theoretical support and guidance for future research.

Modifications to the definition of DMG

The definition of DMG has evolved markedly with the 2016 and 2021 World Health Organization (WHO) CNS tumor classifications, shifting towards molecular pathology-based diagnosis.

Classification and definition in WHO before 2016

DMG, termed diffuse intrinsic pontine glioma (DIPG) before 2016, was first defined by Wilfred Harris in 1926 for its pontine location and histological features (13,14), predominantly affecting children aged 5–10 with a dismal prognosis (median OS less than a year and five-year survival rate <1%) (15). Traditional classification could not distinguish subtypes or elucidate molecular mechanisms (16).

The 2016 WHO classification introduced molecular diagnostics, defining DMG by the H3K27M histone mutation, specifically the K27M mutation in the H3F3A, HIST1H3B and HIST1H3C genes (17,18). This mutation disrupts lysine 27 methylation of histone H3, altering gene regulation and driving tumorigenesis (19), thereby enhancing diagnostic precision and differentiation.

Updates in the 2021 WHO classification

The 2021 WHO classification renamed ‘Diffuse Midline Glioma, H3K27M-mutant’ to ‘Diffuse Midline Glioma, H3 K27-altered’ to encompass broader molecular changes (20), including EZHIP overexpression (21,22), which reduces H3K27me3 levels, promoting invasiveness and therapy resistance (23). These updates improved diagnostic accuracy, highlighted tumor heterogeneity and facilitated tailored, patient-specific treatment strategies to enhance outcomes (24).

Significance of histones in cancer

Definition and functions of histone and oncohistones

Histones are the primary structural proteins of chromatin, packaging DNA into nucleosomes comprising two copies each of H2A, H2B, H3 and H4. Beyond DNA condensation, histones regulate gene expression through post-translational modifications such as methylation, acetylation, phosphorylation and ubiquitination.

Occurring on lysine residues such as H3K4, H3K27 and H4K20, methylation can activate or repress transcription based on its site and extent (mono-, di-, or trimethylation) (25). This process is dynamically regulated by histone methyltransferases (such as SETD1A, which activates transcription via H3K4 methylation) and histone demethylases (such as KDM1A, which represses transcription by demethylating H3K4me2/1) (26). Catalyzed by histone acetyltransferases (HATs) such as p300/CBP, acetylation neutralizes the positive charge of lysine, reducing the electrostatic interaction of histones with the negatively charged DNA (27), loosening chromatin and facilitating transcription. Phosphorylation is closely associated with cell cycle regulation, DNA damage response and transcriptional activation (28,29). Modifications, such as H2AX serine 139 phosphorylation, signal DNA damage, recruit repair proteins and coordinate cell cycle responses (30). Ubiquitination, primarily on H2A and H2B, affects chromatin structure and gene regulation, depending on the site and mono- or poly-ubiquitination status (31).

Oncohistones, a subset of histones predominantly observed in H3 (32,33), drive tumorigenesis and cancer progression by disrupting chromatin structure (34). Mutations alter key modifications such as H3K27 acetylation, which activates oncogenes such as MYC and epidermal growth factor receptor (EGFR) (35) and H3K27me, which silences tumor suppressors by compacting chromatin and blocking transcription factor access. Elevated H3K27me levels are linked to silenced tumor suppressors in breast, liver and lung cancers (36–38). These changes promote tumor proliferation, invasiveness and resistance, making oncohistones pivotal therapeutic targets.

Systemic effects of histone modifications

Histone modifications orchestrate key pathways such as PI3K/AKT/mammalian target of rapamycin (mTOR) and MAPK/ERK (39,40), influencing tumor proliferation, apoptosis and drug resistance (41). Aberrant modifications disrupt gene networks, suppress immune responses and drive metastasis, as seen with elevated HIST1H2BJ activating MMP9 and VEGF in breast cancer brain metastases (42,43). These changes highlight the critical role of histone modifications in cancer progression and metastasis.

H3K27M mutation in the development of DMG

Diffuse midline glioma is defined by the H3K27M oncohistone, which disrupts methylation at histone H3 lysine 27 (H3K27). This missense substitution (lysine-to-methionine) occurs in H3.1 or H3.3 variants and is associated with a profound reduction of H3K27me3, a key epigenetic mark of transcriptional repression. The resulting epigenetic reprogramming reshapes chromatin accessibility and transcriptional programs, thereby promoting aggressive phenotypes, including uncontrolled proliferation, invasion and treatment resistance (44) (Fig. 1).

H3K27M mutation in diffuse midline
glioma. The H3K27M mutation markedly reduces H3K27me3 levels by
inhibiting the EZH2 methyltransferase activity of the PRC2 complex,
leading to disrupted methylation regulation of tumor suppressor
genes and inducing widespread transcriptional abnormalities. This
mutation promotes uncontrolled cell cycle progression, exemplified
by dysregulation of the CDK4/6-p16 pathway and enhances tumor cell
proliferation and invasion through the activation of key signaling
pathways, including MAPK/ERK, PI3K/AKT/mTOR and JAK/STAT. The
H3K27M mutation also induces DNA repair deficiencies by impairing
the homologous recombination repair and base excision repair
pathways. It also affects the cellular stress response by altering
the ATR/ATM kinase pathway, compromising the cell's ability to
manage DNA damage. These defects exacerbate genomic instability and
transcriptional dysregulation, further contributing to tumor
progression. The H3K27M mutation alters m6A methylation and FTO
activity, making DMG cells sensitive to FTO inhibition, which
disrupts cell cycle gene expression and induces apoptosis.
Furthermore, the CHD2/FOSL1 axis plays a crucial role in abnormal
synaptic interactions between neurons and tumor cells, enhancing
tumor growth and invasion by promoting excessive neuronal-tumor
connectivity. Figure created using BioRender.com. H3K27, histone H3
lysine 27, H3K27me3, H3K27 trimethylation; PRC2, polycomb
repressive complex 2; mTOR, mammalian target of rapamycin; FTO,
fat-mass- and obesity-associated protein; DMG, diffuse midline
gliomas.

Figure 1.

H3K27M mutation in diffuse midline glioma. The H3K27M mutation markedly reduces H3K27me3 levels by inhibiting the EZH2 methyltransferase activity of the PRC2 complex, leading to disrupted methylation regulation of tumor suppressor genes and inducing widespread transcriptional abnormalities. This mutation promotes uncontrolled cell cycle progression, exemplified by dysregulation of the CDK4/6-p16 pathway and enhances tumor cell proliferation and invasion through the activation of key signaling pathways, including MAPK/ERK, PI3K/AKT/mTOR and JAK/STAT. The H3K27M mutation also induces DNA repair deficiencies by impairing the homologous recombination repair and base excision repair pathways. It also affects the cellular stress response by altering the ATR/ATM kinase pathway, compromising the cell's ability to manage DNA damage. These defects exacerbate genomic instability and transcriptional dysregulation, further contributing to tumor progression. The H3K27M mutation alters m6A methylation and FTO activity, making DMG cells sensitive to FTO inhibition, which disrupts cell cycle gene expression and induces apoptosis. Furthermore, the CHD2/FOSL1 axis plays a crucial role in abnormal synaptic interactions between neurons and tumor cells, enhancing tumor growth and invasion by promoting excessive neuronal-tumor connectivity. Figure created using BioRender.com. H3K27, histone H3 lysine 27, H3K27me3, H3K27 trimethylation; PRC2, polycomb repressive complex 2; mTOR, mammalian target of rapamycin; FTO, fat-mass- and obesity-associated protein; DMG, diffuse midline gliomas.

Inhibition of H3K27me3 by H3K27M

H3K27 trimethylation is primarily catalyzed by the EZH2 SET domain within the Polycomb Repressive Complex 2 (PRC2), which maintains Polycomb-mediated transcriptional repression programs involved in lineage control and proliferation. Structural and biochemical studies (45) show that the H3K27M oncohistone engages the PRC2 catalytic groove: the substituted methionine inserts into the lysine-access channel of the EZH2 SET domain, thereby competitively inhibiting PRC2 methyltransferase activity and exhibiting markedly higher affinity than the corresponding wild-type H3K27 peptide [equilibrium dissociation constant (Kd) ~3.3±0.4 µM for H3K27M compared with ~52±12 µM for H3K27]. This inhibitory mode provides a mechanistic basis for the global reduction and redistribution of H3K27me3/PRC2 and the ensuing transcriptional reprogramming observed in pediatric H3K27-altered high-grade gliomas (44).

The suppression of H3K27me3 by H3K27M alters gene expression and activates multiple signaling pathways essential for cell proliferation, survival and invasion. In the PI3K/AKT/mTOR axis, growth-factor/RTK inputs activate PI3K and generate PIP3, recruiting and activating AKT, which phosphorylates downstream effectors to promote cell-cycle progression and survival while suppressing apoptosis (39); mTOR further sustains biosynthetic and metabolic programs that facilitate rapid tumor expansion (46).

In parallel, aberrant activation of the JAK/STAT pathway can reinforce proliferative transcriptional programs and contribute to immune-evasive phenotypes within the tumor microenvironment, as JAK-dependent STAT phosphorylation promotes nuclear transcription of genes linked to growth and immunoregulation (44,47).

Moreover, MAPK/ERK signaling, often downstream of RTKs, can be potentiated, enabling RAS-RAF-MEK-ERK propagation and ERK-driven nuclear transcriptional outputs that promote proliferation and therapy resistance (48).

Extensive changes in DNA methylation

The H3K27M mutation disrupts histone methylation and induces widespread changes in DNA methylation. Research by Vanan et al (49) and Graham et al (50) revealed that reduced H3K27me3 in H3K27M mutant cells extends across the genome, altering DNA methylation beyond the sites of mutant histone localization. This indicates that the mutation indirectly affects the regulation of numerous genes.

Further studies by Paine et al (51) demonstrated significant DNA methylation changes, particularly in the promoter regions of tumor suppressor genes such as TP53 and RB1, leading to their silencing and facilitating the malignant progression of tumors.

Typically, the TET family of enzymes (TET1, TET2 and TET3) oxidizes 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5-hmC) in a process known as DNA demethylation (52). However, the H3K27M mutation inhibits TET enzyme activity by disrupting chromatin-associated factors, reducing TET recruitment and markedly decreasing 5-hmC levels. This results in the accumulation of DNA methylation, particularly in the promoter regions of tumor suppressor genes (53,54), creating an epigenetic landscape that promotes DMG invasiveness and proliferation.

Regulation of m6A RNA methylation

Recent research (55) reveals that H3K27M-mutant DMG exhibits unique m6A RNA methylation patterns, with m6A marks enriched on key cell cycle genes. The m6A demethylase fat-mass- and obesity-associated protein (FTO) is critical for DMG cell proliferation; inhibition of FTO (such as by FB23-2) increases m6A levels, disrupts cell cycle progression, reduces tumor cell survival and induces apoptosis. These effects are partly mediated by the m6A reader YTHDF2, which is highly expressed in DMG and promotes degradation of cell cycle transcripts. Targeting FTO-driven m6A dynamics thus represents a promising therapeutic strategy for DMG.

Cell cycle regulation by H3K27M mutation

The H3K27M mutation disrupts cell cycle regulation by inhibiting the activity of the PRC2 complex and silencing tumor suppressor genes such as RB1 and CDKN2A. This leads to the loss of G1/S checkpoint control, allowing cells to bypass critical regulatory mechanisms and proliferate uncontrollably (56) (Fig. 1).

Williams et al (57) highlighted the pivotal role of the p16 protein, encoded by CDKN2A, in maintaining cell cycle control by inhibiting CDK4/6 activity and preventing the transition from the G1 to S phase. In H3K27M mutant tumor cells, the loss of p16 function results in cell cycle imbalance and accelerated tumor growth.

H3K27M mutation inhibits DNA repair

The H3K27M mutation additionally impairs DNA repair processes, exacerbating genomic instability. In normal cells, DNA damage is repaired to preserve genomic integrity. However, in H3K27M mutant cells, the capacity for DNA repair, particularly in the homologous recombination repair and base excision repair pathways, is markedly compromised (Fig. 1). The expression of essential DNA repair genes, including BRCA1, is notably diminished, rendering them prone to accumulating genetic mutations and facilitating tumor progression (58). Furthermore, H3K27M rewires the DNA damage response (DDR) and checkpoint circuitry (including ATM/ATR-linked pathways), which can contribute to treatment resistance but also creates actionable vulnerabilities for DDR-targeted radio sensitization strategies (59).

Neuron-tumor cell interactions

Beyond epigenetic changes, the H3K27M mutation promotes tumor expansion by regulating neuron-tumor interactions (Fig. 1). Chromodomain-Helicase-DNA-binding protein 2 (CHD2) plays a crucial role in chromatin remodeling within the neuronal-tumor microenvironment, regulating genes essential for synaptic formation, neuronal excitability and nerve transmission, such as SYP and PSD95, which maintain synaptic integrity (60).

In H3K27M mutant cells, CHD2 remodels chromatin accessibility and drives an axon-guidance/synaptic transcriptional program; mechanistically, a FOSL1-CHD2 regulatory axis promotes neuron-glioma interactions and neuron-induced tumor proliferation (60). Upregulation of axon-guidance and synaptic gene programs strengthens neuron-glioma interactions and neuron-induced proliferation in H3K27-altered DMG.

Other histone modifications in DMG

Beyond the dysregulation of H3K27 trimethylation by the H3K27M mutation, other histone modifications markedly contribute to DMG progression. These modifications regulate chromatin structure and gene expression, driving tumor cell proliferation, differentiation and invasion (61). Critical histone modifications and their roles in DMG are outlined below.

H3K27ac

H3K27ac, the acetylation of lysine 27 on histone H3, is associated with active chromatin states, promoting gene transcription by maintaining chromatin openness, predominantly at promoters and enhancers.

In DMG, the H3K27M mutation indirectly increases H3K27ac by inhibiting H3K27me3, activating oncogenes such as MYC and PDGFRA, which drive tumor growth. Additionally, this upregulation keeps chromatin at promoter and enhancer regions open, facilitating transcription factors such as SP1 and AP-1 binding (62). Menez et al (56) demonstrated that HATs such as p300 and CBP reinforce this open state, creating a positive feedback loop that amplifies the expression of genes promoting proliferation, survival and invasion.

Mota et al (63) found that the SWI/SNF chromatin remodeling complex, particularly the BAF subtype, contributed to H3K27ac accumulation. The antagonistic interaction between BAF and PRC2 enhances the effects of H3K27ac. Targeting this pathway with AU-15330, which is a PROTAC compound that degrades SMARCA4, SMARCA2 and PBRM1, effectively reduces H3K27ac levels, impairing chromatin openness and inhibiting tumor proliferation.

H3K9me3

H3K9me3 is a repressive histone mark enriched in heterochromatin regions, suppressing gene transcription through chromatin compaction. In DMG, altered H3K9me3 levels are associated with silencing tumor suppressor genes.

Xin et al (64) revealed that the histone methyltransferase SUV39H1 generates H3K9me3 marks, recruiting protein complexes to compact chromatin and repress tumor suppressor genes such as CDKN1A, CDKN2B and CDKN2D, thereby promoting tumor cell proliferation. The fungal metabolite Chaetocin, a SUV39H1 inhibitor, reduces H3K9me3 levels, which restores tumor suppressor gene expression and induces apoptosis in DMG cells.

Notably, when H3K9me3 levels are inhibited, DMG cells activate compensatory pathways such as the dopamine receptor D2 (DRD2) pathway to sustain tumor growth. A combination therapy of Chaetocin and DRD2 antagonists (such as ONC201) has been shown to enhance anti-tumor effects and reduce treatment resistance (64).

H3K36me3

H3K36me3 is an activating histone mark involved in transcriptional elongation and DNA repair. The H3.3G34R/V mutation reduces H3K36me3 levels (51), disrupting transcriptional elongation and mismatch repair, contributing to a mild ‘hyper-mutator’ phenotype by impairing DNA repair mechanisms (65).

The loss of H3K36me3 compromises genomic stability, allowing the accumulation of mutations that drive tumorigenesis. These findings highlight the essential role of H3K36me3 in maintaining cellular function and its dysregulation as a key factor in cancer progression.

Given the pivotal role of these epigenetic modifications in DMG pathogenesis, targeting these epigenetic alterations presents a promising avenue for developing novel therapeutic strategies to combat DMG.

Current treatment regimens

Local treatments, primarily encompassing surgery and radiotherapy, are central to managing DMG. However, the invasive nature of DMG and its midline location limit their effectiveness, underscoring the need for more effective therapeutic options.

Surgical treatments

Surgery for DMG aims to obtain tissue for diagnosis and molecular analysis, as its midline location makes complete resection highly risky. Biopsies, particularly stereotactic ones, highlighted in CPN Paris 2011, are vital for identifying mutations such as H3K27M, enabling targeted therapies (66). Advances in minimally invasive techniques and molecular profiling have made biopsy a key clinical tool, offering critical insights with minimized risks (67).

A multicenter study revealed no survival benefit from resection over biopsy in H3K27M-mutant DMG patients, with OS at 12 months for resection and 11 months for biopsy (68). However, resection was associated with longer ICU stays and higher neurological deficits. Moreover, the surgical intervention did not improve the Karnofsky Performance Status (68,69). Similarly, a systematic review reported a 13.02% complication rate for stereotactic biopsy, with no biopsy-related deaths and a diagnostic success rate of 97.4% (69). While biopsies do not markedly extend survival, they provide invaluable molecular data for personalized therapies.

Radiotherapies
External beam radiotherapy (EBRT)

EBRT remains the standard treatment for DMG, delivering 54–59.4 Gy over 30–33 sessions (70). While it temporarily alleviates symptoms and delays progression for 6–12 months, recurrence is almost inevitable within a year, with a median OS of 8–12 months (71).

Hyper-fractionated radiotherapy and hypo-fractionated radiotherapy

Alternative approaches have been tested, such as hyper-fractionated (e.g., 70.2 Gy in smaller fractions) and hypo-fractionated (e.g., 39 Gy in larger fractions) radiotherapy, but have shown no significant survival benefits. Median survival remains 7.8–8.5 months (71,72), with logistical benefits being the primary advantage.

Proton radiotherapy

Proton therapy minimizes damage to the healthy brain, particularly in pediatric patients, but does not markedly improve long-term survival. Re-radiation after recurrence offers limited benefits and poses a higher risk of radiation-induced damage (73), such as neurological and cognitive impairments (74,75).

Limitations of radiotherapy

Despite temporary relief, tumor progression is inevitable, with progression-free survival (PFS) of only 6–8 months and a 5-year survival rate <1% (76,77). Combining radiotherapy with agents such as Bromodomain and Extra-Terminal motif (BET) inhibitors or epigenetic drugs (such as chitosan) shows potential in augmenting its tumor-killing effects by disrupting DNA repair (66,78).

Chemotherapies

Chemotherapy serves as a complementary treatment for DMG, largely as an adjunct to radiotherapy when resection is not feasible. However, across prospective and registry-era experiences, conventional cytotoxic chemotherapy has not produced a consistent survival benefit beyond radiotherapy alone and its use is increasingly framed within clinical trials or rational combinations rather than as stand-alone escalation (Table I).

Table I.

Summary of clinical trials on chemotherapy, targeted therapy and immunotherapy in the treatment of DMG.

Table I.

Summary of clinical trials on chemotherapy, targeted therapy and immunotherapy in the treatment of DMG.

First author/s, yearTreatment typeDrugTargetsPatients (n)PFS (months)mOS (months)Other outcomes(Refs.)
Bailey et al, 2013ChemotherapyTemozolomide/43/9.51-year OS ~35%; 2-year OS ~17%(79)
Chassot et al, 2013 Temozolomide 227.511.7 (80)
Massimino et al, 2008 Cisplatin + Etoposide 627/1-year OS: 45%(87)
Ono et al, 2021 Cisplatin/Etoposide 1//OS >6 years(89)
Veldhuijzen van Zanten et al, 2017 Gemcitabine 94.88.7 (90)
Kilburn et al, 2018 Capecitabine 44//1-year PFS rate: 7.2%(91)
Krystal et al, 2024 Irinotecan + Bevacizumab + Mebendazole 104.711.4 (93)
Lin et al, 2024 Cyclophosphamide + GD2.CART 11//7/11 neurological improvement(95)
Venneti et al, 2023Targeted TherapiesONC-201ClpP303.49.3 (100)
Arrillaga-Romany et al, 2024 ONC-201ClpP477// (101)
Su et al, 2022 VorinostatHDAC79//1-year PFS rate: 5.85%; 1-year OS rate:39.2%(105)
Mueller et al, 2023 PanobinostatHDAC7726.1 (104)
Neth et al, 2022 PanobinostatHDAC101942 (106)
DeWire et al, 2020 RibociclibCDK4/610/16.1 (111)
DeWire et al, 2022 Ribociclib + EverolimusCDK4/6 + mTOR19/13.9 (112)
Fleischhack et al, 2019 NimotuzumabEGFR425.89.4 (114)
Liu et al, 2025 NimotuzumabEGFR487.810.5 (118)
Majzner et al, 2022ImmunotherapyGD2-CAR T/4//3/4 Tumor volume reduction or stabilization(120)
Lin et al, 2024 C7R-GD2.CART 11//Feasibility; on-target neuroinflammation/TIAN and CRS(95)
Vitanza et al, 2025 B7-H3 CAR-T 21//Repetitive ICV dosing feasible; DLT reported (intratumoral hemorrhage)(122)
Pérez-Larraya et al, 2022 DNX-2401 12/17.8 (124)
Grassl et al, 2023 H3K27M-vac 86.212.8 (125)
Mueller et al, 2020 H3.3K27M peptide vaccine 29 (A=19; B=10)/16.1 vs. 9.8 (128)

[i] PFS, progression-free survival; mOS, median overall survival; ClpP, caseinolytic protease P; HDAC, histone deacetylase; mTOR, mechanistic target of rapamycin; EGFR, epidermal growth factor receptor; TIAN, tumor inflammation-associated neurotoxicity; CRS, cytokine release syndrome; ICV, intracerebroventricular; DLT, dose-limiting toxicity.

Temozolomide

TMZ is an oral alkylating agent whose key cytotoxic lesion is O6-methylguanine, which can trigger replication-associated mismatch repair-dependent DNA damage and apoptosis. In a UK phase II study evaluating dose-dense TMZ with standard radiotherapy for classical DIPG/DMG, median OS was 9.5 months, with 1-year OS of 35% and the regimen did not demonstrate a survival benefit over radiotherapy alone; hematologic toxicity led to treatment withdrawal and frequent dose reductions (79) (Table I). Similarly, radiotherapy with concurrent and adjuvant TMZ yielded median OS of 11.7 months and 1-year OS of 50%, but no significant improvement in outcome and higher hematologic toxicity compared with radiotherapy alone (80). A major biological constraint is O6-methylguanine-DNA methyltransferase (MGMT)-mediated repair of TMZ-induced O6 lesions: H3K27M-altered DMG frequently shows lack of MGMT promoter methylation and increased MGMT expression, which is associated with TMZ resistance in DMG models (81). Accordingly, pharmacologic MGMT inhibition, such as O6-benzylguanine, has been limited by increased hematologic toxicity and often necessitates substantial TMZ dose reduction, constraining clinical utility (82,83). Studies (84–86) demonstrate that BET inhibitors, as well as other epigenetic modulators, including histone deacetylase (HDAC) and EZH2 inhibitors, can sensitize DMG cells to TMZ, with preclinical evidence supporting synergistic antitumor effects when combined.

Cisplatin and etoposide

Cisplatin primarily induces cytotoxicity by forming DNA cross-links, whereas etoposide inhibits topoisomerase II and triggers DNA double-strand breaks; together they provide a rationale for combination regimens that converge on DNA-damage-driven apoptosis.

In a mono-institutional 20-year experience of pediatric diffuse pontine gliomas treated with multi-agent therapy incorporating cisplatin/etoposide (plus isotretinoin and others), the reported median survival was 7 months and the 1-year survival rate was 45% (87). Clinically, limited benefit is assumed to be related in part to inadequate CNS drug exposure under an intact/tight blood-brain barrier (BBB) in DMG (88), although rare long survivors exist. For example, a pineal-region DMG case with >6-year survival after multimodal management in which initial carboplatin-etoposide was ineffective but extensive resection, high-dose radiotherapy and bevacizumab-based maintenance was associated with durable control (89).

Gemcitabine

Gemcitabine is a cytidine analog that inhibits DNA synthesis via DNA polymerase and ribonucleotide reductase and has been explored as a radiosensitizer in DMG (9,90).

In a phase I/II trial of weekly gemcitabine with radiotherapy, treatment was safe and well tolerated, with PFS 4.8 months and median OS 8.7 months (90); Quality of life tended to improve, but survival was not superior to historical controls, supporting mainly palliative value.

Capecitabine

Capecitabine is an oral prodrug of 5-fluorouracil; because radiation can induce thymidine phosphorylase, capecitabine has been tested as a potential RT-sensitizing strategy in DMG (91). In the PBTC phase Il study (91), capecitabine + RT was tolerable, but outcomes were not improved: 1-year PFS 7.21 vs. 15.59% in the historical control (P=0.007) and no OS difference (P=0.30), leading to the conclusion that capecitabine did not improve prognosis in newly diagnosed DMG.

Irinotecan

Irinotecan is a topoisomerase I inhibitor that stabilizes the cleavage complex and induces lethal DNA strand breaks, thereby suppressing tumor proliferation (92). In a phase I trial, Krystal et al (93) evaluated irinotecan plus bevacizumab and mebendazole in 10 pediatric/AYA HGG patients, including 7 DMG, reporting no dose limiting toxicities and a PFS and OS of 4.7 and 11.4 months from treatment initiation; the overall response rate was 33% (two PRs and one CR sustained for 10 months).

Cyclophosphamide

Cyclophosphamide is an alkylating pro-drug that generates phosphoramide mustard, producing DNA cross-links and triggering apoptosis (94).

In a phase I trial of intravenous GD2. CAR T cells, cyclophosphamide (with fludarabine) was used for standard lymphodepletion in 11 patients and therapy was delivered without dose-limiting toxicity. The C7R-GD2.CAR T cohort developed grade 1 TIAN in 7/8 and achieved transient neurologic improvement (2 to >12 months), with partial responses in 2/7 DMG patients by iRANO (95).

Overall, conventional chemotherapies have shown limited and largely non-practice-changing benefit in DMG. Future efforts should prioritize rational combinations (such as chemo with targeted/epigenetic or immunotherapies) and trial designs that emphasize clinically meaningful endpoints, including neurologic function and quality of life.

Targeted therapies

Targeted therapy for diffuse midline glioma aims to exploit actionable biological dependencies that sustain tumor growth. Current strategies include mitochondrial stress-based approaches centered on caseinolytic protease P (ClpP)/imipridones such as ONC201, epigenetic modulation such as HDAC inhibitors, panobinostat for epigenetic regulation and combination therapies targeting CDK4/6 and mTOR and receptor-directed interventions focusing on EGFR inhibition. These modalities are being evaluated to counteract core metabolic/epigenetic vulnerabilities and aberrant signaling programs in DMG (Fig. 2; Table I).

Mechanisms of targeted therapy in
diffuse midline glioma. (A) ClpP agonist ONC-201 hyperactivates
mitochondrial protease ClpP, leading to respiratory chain protein
degradation, electron transport chain disruption, OXPHOS inhibition
and reduced ATP production. (B) HDAC inhibitor panobinostat
restores p53 function through MDM2/MDM4 modulation and enhanced p53
acetylation, upregulating pro-apoptotic Bax while downregulating
anti-apoptotic Bcl-2 to induce tumor cell apoptosis. (C) CDK4/6
inhibitor ribociclib blocks CDK4/6-cyclin D interaction, inducing
G1 arrest; mTOR inhibitor everolimus suppresses
PI3K/AKT/mTOR signaling to inhibit proliferation and metabolism.
(D) Anti-EGFR antibody nimotuzumab blocks EGF/TGF-α binding,
inhibiting downstream tyrosine kinase signaling and
HIF-1α/VEGF-mediated angiogenesis. Figure created using
BioRender.com. ClpP, caseinolytic protease P; HDAC, histone
deacetylase; mTOR, mammalian target of rapamycin; EGFR, epidermal
growth factor receptor; HIF-1α, hypoxia-inducible factor
1-alpha.

Figure 2.

Mechanisms of targeted therapy in diffuse midline glioma. (A) ClpP agonist ONC-201 hyperactivates mitochondrial protease ClpP, leading to respiratory chain protein degradation, electron transport chain disruption, OXPHOS inhibition and reduced ATP production. (B) HDAC inhibitor panobinostat restores p53 function through MDM2/MDM4 modulation and enhanced p53 acetylation, upregulating pro-apoptotic Bax while downregulating anti-apoptotic Bcl-2 to induce tumor cell apoptosis. (C) CDK4/6 inhibitor ribociclib blocks CDK4/6-cyclin D interaction, inducing G1 arrest; mTOR inhibitor everolimus suppresses PI3K/AKT/mTOR signaling to inhibit proliferation and metabolism. (D) Anti-EGFR antibody nimotuzumab blocks EGF/TGF-α binding, inhibiting downstream tyrosine kinase signaling and HIF-1α/VEGF-mediated angiogenesis. Figure created using BioRender.com. ClpP, caseinolytic protease P; HDAC, histone deacetylase; mTOR, mammalian target of rapamycin; EGFR, epidermal growth factor receptor; HIF-1α, hypoxia-inducible factor 1-alpha.

ClpP

ClpP, a mitochondrial serine protease, forms the ClpXP complex to maintain mitochondrial proteostasis. Pharmacologic hyperactivation of ClpP can drive excessive proteolysis of respiratory-chain proteins, leading to electron transport chain disruption, OXPHOS inhibition, ATP depletion and apoptotic cell death (96).

ONC201, an imipridone, acts as an allosteric ClpP agonist and antagonizes DRD2. In H3K27M-altered DMG models, ONC201-ClpP engagement promotes degradation of key respiratory chain components, collapses mitochondrial bioenergetics (ETC/OXPHOS) and suppresses tumor energy metabolism-features that match the present study's mechanistic schematic (96,97) (Fig. 2A). Downstream, ONC201 induces an ATF4/CHOP-linked integrated stress response and can promote DR5/TRAIL-mediated apoptosis (98), with reported modulation of Akt/ERK signaling consistent with a pro-apoptotic shift (99).

Clinically, integrated analysis of ONC201-treated H3K27M-mutant DMG reported longer survival when initiated after radiotherapy but before recurrence (median OS 21.7 months; PFS 7.3 months) compared with initiation after recurrence (median OS 9.3 months; PFS 3.4 months), with molecular correlates including increased 2-hydroxyglutarate and partial restoration of H3K27me3 (100). A randomized, double-blind phase 3 trial (101) is ongoing to evaluate ONC201 as post-radiotherapy maintenance therapy in newly diagnosed H3 K27M-mutant diffuse glioma, with OS and PFS as primary endpoints.

HDAC

HDAC inhibitors reprogram chromatin by increasing histone acetylation and can reactivate the MDM2/MDM4-p53 axis, in part by reducing MDM2/MDM4 repression and enhancing p53 acetylation/transcriptional activity, thereby promoting apoptosis (102). Downstream, they upregulate pro-apoptotic genes such as Bax while downregulate anti-apoptotic genes such as Bcl-2, activating the intrinsic apoptotic pathway (103) (Fig. 2B).

Panobinostat is a multi-HDAC inhibitor with strong preclinical rationale in DMG, but systemic delivery is constrained by exposure/toxicity and uncertain BBB penetration, motivating local delivery approaches (104).

In a phase I/II trial by Su et al (105), panobinostat combined with radiotherapy and voriostat, followed by maintenance, was well-tolerated but showed limited survival benefits, with a 1-year PFS of 5.85% and OS of 39.2%. By contrast, PNOC015 using repeat CED of MTX110 reported median OS of 26.1 months and median PFS 7 months, supporting intra-tumoral HDAC inhibition with reduced systemic exposure (104). In adults, compassionate-use panobinostat showed good tolerability with median OS 42 months and median PFS 19 months in a small series, suggesting potential context-dependence that requires prospective validation (106).

Cyclin-dependent kinases 4 and 6 (CDK4/6) + mTOR

CDK4/6 regulate the G1-to-S phase transition by phosphorylating retinoblastoma (RB) protein via cyclin D, activating E2F transcription factors and driving cell cycle progression (107,108). The mTOR in the PI3K/Akt pathway governs metabolism, protein synthesis and proliferation, with aberrations frequently observed in DMG, contributing to unchecked tumor proliferation and metabolic dependency (107).

Ribociclib, a CDK4/6 inhibitor, halts cell cycle progression at the G1 phase by disrupting CDK4/6-cyclin D interaction (109) (Fig. 2C), targeting CDKN2A/B deletions and CCND2 amplifications seen in DMG. Meanwhile, everolimus, which is a mTORC1 inhibitor, suppresses tumor proliferation, glycolysis and angiogenesis (110), particularly in tumors with H3.1 mutations linked to PIK3CA or PIK3R1 alterations (40). The combination of ribociclib and everolimus provides dual inhibition of cell cycle and metabolic pathways, enhancing antitumor effects with reduced overlapping toxicity.

A phase I/II study of post-radiotherapy single-agent ribociclib reported feasibility with myelosuppression as the main severe toxicity and a median OS of 16.1 months in DIPG/DMG, reinforcing ongoing interest in rational combinations and biomarker-guided selection (111). As additional supportive evidence for CDK4/6 targeting in this setting, A phase I trial assessed this combination in 19 pediatric DMG patients following radiotherapy, establishing phase II doses of 170 mg/m2 for ribociclib and 1.5 mg/m2 for everolimus (112). The median OS was 13.9 months, with 12-, 24- and 36-month survival rates of 53.3, 38.9 and 38.9%, respectively. These promising results warrant further investigation of this approach.

EGFR

EGFR is a receptor tyrosine kinase that propagates mitogenic and pro-survival signals mainly through the RAS-RAF-MEK-ERK and PI3K-AKT cascades, thereby promoting proliferation, invasion and treatment resistance (113). Additionally, EGFR pathway activity intersects with hypoxia/angiogenic programs, in part via PI3K/AKT-hypoxia-inducible factor 1-alpha (HIF-1α)-VEGF, providing a rationale for EGFR-directed strategies to suppress both tumor growth and angiogenesis (114).

Nimotuzumab, an anti-EGFR monoclonal antibody, blocks EGF/TGF-α-EGFR engagement and thereby attenuates downstream oncogenic signaling; clinically, however, its benefit has been inconsistent (115). It induces G1 phase arrest, preventing tumor cell progression into the S phase (116). Additionally, it destabilizes HIF-1α, suppresses VEGF transcription and reduces angiogenesis, collectively curbing tumor growth and metastasis (117) (Fig. 2D).

A phase III trial in 42 DMG patients evaluated nimotuzumab with radiotherapy, reporting a median PFS of 5.8 months and median OS of 9.4 months (114). One- and two-year survival rates were 33.3 and 4.8%, respectively, with rare serious events including intratumoral bleeding. More recently, adding nimotuzumab to chemoradiation was feasible, with median OS 10.5 months and PFS 7.8 months, but did not meet a prespecified ORR-improvement threshold vs. historical data, supporting continued biomarker-guided optimization rather than routine adoption (118).

Immunotherapies

Diffuse midline glioma is characterized as an immunologically ‘cold tumor’, with low lymphocytic infiltration and a microenvironment dominated by myeloid-lineage populations, which collectively constrains adaptive anti-tumor immunity and limits the efficacy of conventional immunotherapy approaches (119). These features highlight why DMG immunotherapy remains early-stage and why locoregional, antigen-directed strategies have become a central development focus (Table I).

Chimeric antigen receptor (CAR)-T therapy

CAR-T-cell therapy aims to bypass limited endogenous T-cell priming by engineering T cells to recognize DMG-associated surface antigens, most prominently GD2 and B7-H3 (120). Upon antigen engagement, CAR signaling through CD3Z activates canonical T-cell effector programs, such as cytokine production and cytotoxicity, enabling direct killing of antigen-expressing tumor cells.

For GD2 targeting, first-in-human clinical experience demonstrated feasibility and early signals of activity, while highlighting neuroinflammation as an on-target risk that requires proactive monitoring and management (120). More recently, a phase I study of constitutively active IL-7 receptor-engineered GD2 CAR T cells (C7R-GD2.CART), further supported feasibility in H3K27-altered DMG, with tumor-inflammation-associated neurotoxicity and cytokine-release syndrome as key safety considerations and preliminary objective responses observed in a subset (95).

In parallel, B7-H3-directed CAR T cells have advanced through BrainChild-03 using repeated intra-cerebroventricular dosing; early DIPG evaluable cases showed no dose-limiting toxicities and evidence of local immune activation with a durable clinical/radiographic improvement in one patient (121). Updated Arm C results in DIPG expanded these observations by establishing feasibility of repetitive ICV dosing at higher planned dose regimens and providing benchmark survival and neurotoxicity profiles for future multi-site evaluation (122).

Oncolytic virus DNX-2401

DNX-2401 is a conditionally replicating adenovirus designed to preferentially replicate in tumors with RB pathway dysregulation via a Δ24 deletion in E1A and its RGD modification can facilitate integrin-mediated entry (123).

In a phase I study of 12 newly diagnosed DMG/DIPG patients, a single stereotactic intra-tumoral infusion of DNX-2401 (1×1010 or 5×1010 viral particles) followed by radiotherapy achieved 92% disease control and a median OS of 17.8 months; 12- and 18-month OS were 75 and 50%, respectively (124). Correlative analysis supported treatment-associated immune activation, including increased T-cell-related activity, aligning with an oncolytic ‘viro-immunotherapy’ rationale in DMG.

Cancer vaccine

Cancer vaccines aims to induce tumor antigen-specific adaptive immunity. In DMG, the shared clonal neo-antigen H3K27M has enabled peptide-vaccine strategies that predominantly prime mutation-specific CD4+ T-cell responses, with potential downstream support for cytotoxic effector programs and can increase immune infiltration in selected patients (125). Mechanistically, deeper immune profiling in a long-term recovered responder following H3K27M vaccination, identified broad HLA background coverage with both B- and T-cell responses, supporting the concept that vaccine-elicited anti-H3K27M immunity can be durable in selected cases (126,127).

The first-in-human H3K27M peptide vaccine (H3K27M-vac) study enrolled eight adults with H3K27M-altered DMG and showed favorable tolerability with mutation-specific immune responses dominated by CD4+ T cells; reported outcomes included median PFS 6.2 months and median OS 12.8 months (125). In younger patients, PNOC007 evaluated an HLA-restricted H3.3K27M peptide vaccine with adjuvant immunostimulation after radiotherapy and demonstrated immunogenicity with acceptable safety, reporting ~40% (DIPG) and ~39% (non-pontine DMG) 12-month OS benchmarks (128). INTERCEPT H3 is currently evaluating the use of H3K27M-vac concurrently with standard radiotherapy and in combination with the PD-L1 antibody atezolizumab for maintenance after radiotherapy to clarify its safety and immunogenicity (129).

Conclusion

Diffuse midline glioma, particularly the H3K27M-mutated subtype, remains a formidable CNS malignancy due to its unique molecular features and invasiveness. The present review emphasized the pivotal role of histone alterations, especially H3K27M, in reshaping chromatin states and gene expression programs that drive malignant proliferation, invasion and therapeutic resistance. Although surgery and radiotherapy remain foundational, their effect is constrained by anatomical inoperability and diffuse infiltration. Chemotherapy provides only a modest benefit and is frequently limited by resistance. By contrast, emerging targeted approaches, including ONC-201, panobinostat-based strategies and CDK4/6 inhibition combined with mTOR blockade, directly address molecular vulnerabilities, while early immunotherapy studies, including locoregional CAR-T platforms, DNX-2401 virotherapy and H3K27M-directed vaccination, illustrate feasibility and biological activity in selected settings.

Limitations of the current evidence base include small patient numbers, heterogeneous cohorts (age, tumor location, co-alterations and prior therapy) and variable drug-delivery strategies, which collectively limit cross-trial comparability and may inflate apparent signals in single-arm studies. Future progress will require biomarker-guided stratification, rational combination regimens aligned to DMG biology (including radiotherapy-integrated approaches) and delivery-optimized platforms that improve intra-tumoral exposure while maintaining neurological safety. Equally important, standardized response assessment and integrated correlative monitoring (molecular and immune profiling, longitudinal sampling, where feasible) should be embedded into multicenter trials to distinguish durable benefit from transient inflammatory effects and to enable definitive efficacy testing.

Acknowledgements

Not applicable.

Funding

The authors gratefully acknowledge the funding support provided by the National High-Level Hospital Clinical Research Funding (grant no. 2022-PUMCH-B-113), the CAMS Innovation Fund for Medical Sciences (grant no. 2021-12M-1-014) and the National Natural Science Foundation of China (grant no. 82151302), which markedly contributed to the advance of this research.

Availability of data and materials

Not applicable.

Authors' contributions

WBW and WLC performed the literature search, extracted and synthesized data on the epigenetic mechanisms and therapeutic strategies of diffuse midline glioma and drafted the original manuscript. WBW prepared and revised the figures and schematic illustrations and WLC organized the tables. WBM and YW conceived and supervised the overall topic and structure of the present review, provided critical revision of the manuscript for important intellectual content and oversaw the final interpretation of the literature. YW and WBM are the corresponding authors and take primary responsibility for the integrity of the work as a whole. WBW and WLC contributed equally to this work. All authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ostrom QT, Price M, Neff C, Cioffi G, Waite KA, Kruchko C and Barnholtz-Sloan JS: CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2016–2020. Neuro Oncol. 25 (12 Suppl 2):iv1–iv99. 2023. View Article : Google Scholar : PubMed/NCBI

2 

Fonseca A, Afzal S, Bowes L, Crooks B, Larouche V, Jabado N, Perreault S, Johnston DL, Zelcer S, Fleming A, et al: Pontine gliomas a 10-year population-based study: A report from The Canadian Paediatric Brain Tumour Consortium (CPBTC). J Neurooncol. 149:45–54. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Patil N, Kelly ME, Yeboa DN, Buerki RA, Cioffi G, Balaji S, Ostrom QT, Kruchko C and Barnholtz-Sloan JS: Epidemiology of brainstem high-grade gliomas in children and adolescents in the United States, 2000–2017. Neuro Oncol. 23:990–998. 2021. View Article : Google Scholar : PubMed/NCBI

4 

Erker C, Lane A, Chaney B, Leary S, Minturn JE, Bartels U, Packer RJ, Dorris K, Gottardo NG, Warren KE, et al: Characteristics of patients ≥10 years of age with diffuse intrinsic pontine glioma: A report from the International DIPG/DMG registry. Neuro Oncol. 24:141–152. 2022. View Article : Google Scholar : PubMed/NCBI

5 

Hoffman LM, Veldhuijzen van Zanten SEM, Colditz N, Baugh J, Chaney B, Hoffmann M, Lane A, Fuller C, Miles L, Hawkins C, et al: Clinical, radiologic, pathologic, and molecular characteristics of long-term survivors of diffuse intrinsic pontine glioma (DIPG): A collaborative report from the international and European society for pediatric oncology DIPG registries. J Clin Oncol. 36:1963–1972. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Ying Y, Liu X, Li X, Mei N, Ruan Z, Lu Y and Yin B: Distinct MRI characteristics of spinal cord diffuse midline glioma, H3 K27-altered in comparison to spinal cord glioma without H3 K27-alteration and demyelination disorder. Acta Radiol. 65:284–293. 2024. View Article : Google Scholar : PubMed/NCBI

7 

Nafe R, Porto L, Samp PF, You SJ and Hattingen E: Adult-type and Pediatric-type diffuse gliomas what the neuroradiologist should know. Clin Neuroradiol. 33:611–624. 2023. View Article : Google Scholar : PubMed/NCBI

8 

Coleman C, Chen K, Lu A, Seashore E, Stoller S, Davis T, Braunstein S, Gupta N and Mueller S: Interdisciplinary care of children with diffuse midline glioma. Neoplasia. 35:1008512023. View Article : Google Scholar : PubMed/NCBI

9 

Saratsis AM, Knowles T, Petrovic A and Nazarian J: H3K27M mutant glioma: Disease definition and biological underpinnings. Neuro Oncol. 26 (Suppl 2):S92–S100. 2024. View Article : Google Scholar : PubMed/NCBI

10 

Tarapore RS, Arain S, Blaine E, Hsiung A, Melemed AS and Allen JE: Immunohistochemistry detection of Histone H3 K27M mutation in human glioma tissue. Appl Immunohistochem Mol Morphol. 32:96–101. 2024. View Article : Google Scholar : PubMed/NCBI

11 

Duchatel RJ, Jackson ER, Alvaro F, Nixon B, Hondermarck H and Dun MD: Signal transduction in diffuse intrinsic pontine glioma. Proteomics. 19:e18004792019. View Article : Google Scholar : PubMed/NCBI

12 

Testa U, Castelli G and Pelosi E: CAR-T cells in the treatment of nervous system tumors. Cancers (Basel). 16:29132024. View Article : Google Scholar : PubMed/NCBI

13 

Harris W: A case of pontine glioma, with special reference to the paths of gustatory sensation. Proc R Soc Med. 19:1–5. 1926.PubMed/NCBI

14 

Janssens GO, Kramm CM and von Bueren AO: Diffuse intrinsic pontine gliomas (DIPG) at recurrence: Is there a window to test new therapies in some patients? J Neurooncol. 139:5012018. View Article : Google Scholar : PubMed/NCBI

15 

Ward E, DeSantis C, Robbins A, Kohler B and Jemal A: Childhood and adolescent cancer statistics, 2014. CA Cancer J Clin. 64:83–103. 2014.PubMed/NCBI

16 

Gwak HS and Park HJ: Developing chemotherapy for diffuse pontine intrinsic gliomas (DIPG). Crit Rev Oncol Hematol. 120:111–119. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P and Ellison DW: The 2016 World Health Organization classification of tumors of the central nervous system: A summary. Acta Neuropathol. 131:803–820. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Huang TY, Piunti A, Lulla RR, Qi J, Horbinski CM, Tomita T, James CD, Shilatifard A and Saratsis AM: Detection of Histone H3 mutations in cerebrospinal fluid-derived tumor DNA from children with diffuse midline glioma. Acta Neuropathol Commun. 5:282017. View Article : Google Scholar : PubMed/NCBI

19 

Wang Q, Niu W and Pan H: Targeted therapy with anlotinib for a H3K27M mutation diffuse midline glioma patient with PDGFR-alpha mutation: A case report. Acta Neurochir (Wien). 164:2063–2066. 2022. View Article : Google Scholar : PubMed/NCBI

20 

McNamara C, Mankad K, Thust S, Dixon L, Limback-Stanic C, D'Arco F, Jacques TS and Löbel U: 2021 WHO classification of tumours of the central nervous system: A review for the neuroradiologist. Neuroradiology. 64:1919–1950. 2022. View Article : Google Scholar : PubMed/NCBI

21 

Cooney TM, Lubanszky E, Prasad R, Hawkins C and Mueller S: Diffuse midline glioma: review of epigenetics. J Neurooncol. 150:27–34. 2020. View Article : Google Scholar : PubMed/NCBI

22 

Park YW, Vollmuth P, Foltyn-Dumitru M, Sahm F, Ahn SS, Chang JH and Kim SH: The 2021 WHO Classification for Gliomas and implications on imaging diagnosis: Part 2-summary of imaging findings on pediatric-type diffuse high-grade gliomas, pediatric-type diffuse low-grade gliomas, and circumscribed astrocytic gliomas. J Magn Reson Imaging. 58:690–708. 2023. View Article : Google Scholar : PubMed/NCBI

23 

Cassim A, Dun MD, Gallego-Ortega D and Valdes-Mora F: EZHIP's role in diffuse midline glioma: echoes of oncohistones? Trends Cancer. 10:1095–1105. 2024. View Article : Google Scholar : PubMed/NCBI

24 

Hoffman LM, DeWire M, Ryall S, Buczkowicz P, Leach J, Miles L, Ramani A, Brudno M, Kumar SS, Drissi R, et al: Spatial genomic heterogeneity in diffuse intrinsic pontine and midline high-grade glioma: Implications for diagnostic biopsy and targeted therapeutics. Acta Neuropathol Commun. 4:12016. View Article : Google Scholar : PubMed/NCBI

25 

Greer EL and Shi Y: Histone methylation: A dynamic mark in health, disease and inheritance. Nat Rev Genet. 13:343–357. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Hyun K, Jeon J, Park K and Kim J: Writing, erasing and reading histone lysine methylations. Exp Mol Med. 49:e3242017. View Article : Google Scholar : PubMed/NCBI

27 

Shvedunova M and Akhtar A: Modulation of cellular processes by histone and non-histone protein acetylation. Nat Rev Mol Cell Biol. 23:329–349. 2022. View Article : Google Scholar : PubMed/NCBI

28 

An T, Liu Y, Gourguechon S, Wang CC and Li Z: CDK phosphorylation of translation initiation factors couples protein translation with cell-cycle transition. Cell Rep. 25:3204–3214.e5. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Justice JL, Reed TJ, Phelan B, Greco TM, Hutton JE and Cristea IM: DNA-PK and ATM drive phosphorylation signatures that antagonistically regulate cytokine responses to herpesvirus infection or DNA damage. Cell Syst. 15:339–361.e8. 2024. View Article : Google Scholar : PubMed/NCBI

30 

Chen Q, Bian C, Wang X, Liu X, Ahmad Kassab M, Yu Y and Yu X: ADP-ribosylation of histone variant H2AX promotes base excision repair. EMBO J. 40:e1045422021. View Article : Google Scholar : PubMed/NCBI

31 

Mattiroli F and Penengo L: Histone Ubiquitination: An integrative signaling platform in genome stability. Trends Genet. 37:566–581. 2021. View Article : Google Scholar : PubMed/NCBI

32 

Zhang X and Zhang Z: Oncohistone mutations in diffuse intrinsic pontine glioma. Trends Cancer. 5:799–808. 2019. View Article : Google Scholar : PubMed/NCBI

33 

Schwartzentruber J, Korshunov A, Liu XY, Jones DT, Pfaff E, Jacob K, Sturm D, Fontebasso AM, Quang DA, Tönjes M, et al: Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature. 482:226–231. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Qin J, Wen B, Liang Y, Yu W and Li H: Histone modifications and their role in colorectal cancer (review). Pathol Oncol Res. 26:2023–2033. 2020. View Article : Google Scholar : PubMed/NCBI

35 

Caslini C, Hong S, Ban YJ, Chen XS and Ince TA: HDAC7 regulates histone 3 lysine 27 acetylation and transcriptional activity at super-enhancer-associated genes in breast cancer stem cells. Oncogene. 38:6599–6614. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Deng L, Meng T, Chen L, Wei W and Wang P: The role of ubiquitination in tumorigenesis and targeted drug discovery. Signal Transduct Target Ther. 5:112020. View Article : Google Scholar : PubMed/NCBI

37 

Cole AJ, Clifton-Bligh R and Marsh DJ: Histone H2B monoubiquitination: Roles to play in human malignancy. Endocr Relat Cancer. 22:T19–T33. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Chen X, Song N, Matsumoto K, Nanashima A, Nagayasu T, Hayashi T, Ying M, Endo D, Wu Z and Koji T: High expression of trimethylated histone H3 at lysine 27 predicts better prognosis in non-small cell lung cancer. Int J Oncol. 43:1467–1480. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Stitzlein LM, Adams JT, Stitzlein EN, Dudley RW and Chandra J: Current and future therapeutic strategies for high-grade gliomas leveraging the interplay between epigenetic regulators and kinase signaling networks. J Exp Clin Cancer Res. 43:122024. View Article : Google Scholar : PubMed/NCBI

40 

Pearson AD, DuBois SG, Macy ME, de Rojas T, Donoghue M, Weiner S, Knoderer H, Bernardi R, Buenger V, Canaud G, et al: Paediatric strategy forum for medicinal product development of PI3-K, mTOR, AKT and GSK3beta inhibitors in children and adolescents with cancer. Eur J Cancer. 207:1141452024. View Article : Google Scholar : PubMed/NCBI

41 

Yan Y and Zhang J: Mechanisms of tamoxifen resistance: Insight from long non-coding RNAs. Front Oncol. 14:14585882024. View Article : Google Scholar : PubMed/NCBI

42 

Zhang Y, Tapinos N, Lulla R and El-Deiry WS: Dopamine pre-treatment impairs the anti-cancer effect of integrated stress response- and TRAIL pathway-inducing ONC201, ONC206 and ONC212 imipridones in pancreatic, colorectal cancer but not DMG cells. Am J Cancer Res. 14:2453–2464. 2024. View Article : Google Scholar : PubMed/NCBI

43 

Nandy D, Rajam SM and Dutta D: A three layered histone epigenetics in breast cancer metastasis. Cell Biosci. 10:522020. View Article : Google Scholar : PubMed/NCBI

44 

Blasco-Santana L and Colmenero I: Molecular and pathological features of paediatric high-grade gliomas. Int J Mol Sci. 25:84982024. View Article : Google Scholar : PubMed/NCBI

45 

Justin N, Zhang Y, Tarricone C, Martin SR, Chen S, Underwood E, De Marco V, Haire LF, Walker PA, Reinberg D, et al: Structural basis of oncogenic histone H3K27M inhibition of human polycomb repressive complex 2. Nat Commun. 7:113162016. View Article : Google Scholar : PubMed/NCBI

46 

Arakaki AKS, Szulzewsky F, Gilbert MR, Gujral TS and Holland EC: Utilizing preclinical models to develop targeted therapies for rare central nervous system cancers. Neuro Oncol. 23 (23 Suppl 5):S4–S15. 2021. View Article : Google Scholar : PubMed/NCBI

47 

Pal S, Kaplan JP, Nguyen H, Stopka SA, Savani MR, Regan MS, Nguyen QD, Jones KL, Moreau LA, Peng J, et al: A druggable addiction to de novo pyrimidine biosynthesis in diffuse midline glioma. Cancer Cell. 40:957–972.e10. 2022. View Article : Google Scholar : PubMed/NCBI

48 

Tsai JW, Cejas P, Coppola M, Wang DK, Patel S, Wu DW, Arounleut P, Wei X, Zhou N, Syamala S, et al: Abstract 3562: Dissecting mechanisms underlying FOXR2-mediated gliomagenesis in diffuse midline gliomas. Cancer Res. 83:35622023. View Article : Google Scholar : PubMed/NCBI

49 

Vanan MI, Underhill DA and Eisenstat DD: Targeting epigenetic pathways in the treatment of pediatric diffuse (High Grade) gliomas. Neurotherapeutics. 14:274–283. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Graham MS and Mellinghoff IK: Histone-Mutant glioma: Molecular mechanisms, preclinical models, and implications for therapy. Int J Mol Sci. 21:71932020. View Article : Google Scholar : PubMed/NCBI

51 

Paine DZ: Molecular Determinants of Diffuse Midline Glioma of the Pons Vulnerability to the Histone Deacetylase Inhibitor, Quisinostat. Doctoral dissertation. University of Arizona; Tucson, USA: 2023

52 

Xu Y, Wu F, Tan L, Kong L, Xiong L, Deng J, Barbera AJ, Zheng L, Zhang H, Huang S, et al: Genome-wide Regulation of 5hmC, 5mC, and gene expression by Tet1 hydroxylase in mouse embryonic stem cells. Mol Cell. 42:451–464. 2011. View Article : Google Scholar : PubMed/NCBI

53 

Hashizume R: Epigenetic targeted therapy for diffuse intrinsic pontine glioma. Neurol Med Chir (Tokyo). 57:331–342. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Markouli M, Strepkos D, Papavassiliou KA, Papavassiliou AG and Piperi C: Crosstalk of epigenetic and metabolic signaling underpinning glioblastoma pathogenesis. Cancers (Basel). 14:26552022. View Article : Google Scholar : PubMed/NCBI

55 

Ross SE, Holliday H, Tsoli M, Ziegler DS and Dinger ME: Abstract B015: RNA N6-methyladenosine (m6A) as a therapeutic target in Diffuse Midline Glioma (DMG). Cancer Res. 84:B015. 2024. View Article : Google Scholar

56 

Menez V, Kergrohen T, Shasha T, Silva-Evangelista C, Le Dret L, Auffret L, Subecz C, Lancien M, Ajlil Y, Vilchis IS, et al: VRK3 depletion induces cell cycle arrest and metabolic reprogramming of pontine diffuse midline glioma-H3K27 altered cells. Front Oncol. 13:12293122023. View Article : Google Scholar : PubMed/NCBI

57 

Williams EA, Brastianos PK, Wakimoto H, Zolal A, Filbin MG, Cahill DP, Santagata S and Juratli TA: A comprehensive genomic study of 390 H3F3A-mutant pediatric and adult diffuse high-grade gliomas, CNS WHO grade 4. Acta Neuropathol. 146:515–525. 2023. View Article : Google Scholar : PubMed/NCBI

58 

Yan Y: Abstract 1719: Targeting H3.3 serine 28 phosphorylation as a novel therapeutic strategy for diffuse midline glioma. Cancer Res. 84:1719. 2024. View Article : Google Scholar : PubMed/NCBI

59 

Mangoli A, Valentine V, Maingi SM, Wu SR, Liu HQ, Aksu M, Jain V, Foreman BE, Regal JA, Weidenhammer LB, et al: Disruption of ataxia telangiectasia-mutated kinase enhances radiation therapy efficacy in spatially directed diffuse midline glioma models. J Clin Invest. 135:e1793952025. View Article : Google Scholar : PubMed/NCBI

60 

Zhang X, Duan S, Apostolou PE, Wu X, Watanabe J, Gallitto M, Barron T, Taylor KR, Woo PJ, Hua X, et al: CHD2 Regulates Neuron-glioma interactions in pediatric glioma. Cancer Discov. 14:1732–1754. 2024. View Article : Google Scholar : PubMed/NCBI

61 

Diaz AK and Baker SJ: The genetic signatures of pediatric high-grade glioma: No longer a one-act play. Semin Radiat Oncol. 24:240–247. 2014. View Article : Google Scholar : PubMed/NCBI

62 

Sharma M, Barravecchia I, Magnuson B, Ferris SF, Apfelbaum A, Mbah NE, Cruz J, Krishnamoorthy V, Teis R, Kauss M, et al: Histone H3 K27M-mediated regulation of cancer cell stemness and differentiation in diffuse midline glioma. Neoplasia. 44:1009312023. View Article : Google Scholar : PubMed/NCBI

63 

Mota M, Sweha SR, Pun M, Natarajan SK, Ding Y, Chung C, Hawes D, Yang F, Judkins AR, Samajdar S, et al: Targeting SWI/SNF ATPases in H3.3K27M diffuse intrinsic pontine gliomas. Proc Natl Acad Sci USA. 120:e22211751202023. View Article : Google Scholar : PubMed/NCBI

64 

Xin DE, Liao Y, Rao R, Ogurek S, Sengupta S, Xin M, Bayat AE, Seibel WL, Graham RT, Koschmann C and Lu QR: Chaetocin-mediated SUV39H1 inhibition targets stemness and oncogenic networks of diffuse midline gliomas and synergizes with ONC201. Neuro Oncol. 26:735–748. 2024. View Article : Google Scholar : PubMed/NCBI

65 

Fang J, Huang Y, Mao G, Yang S, Rennert G, Gu L, Li H and Li GM: Cancer-driving H3G34V/R/D mutations block H3K36 methylation and H3K36me3-MutSα interaction. Proc Natl Acad Sci USA. 115:9598–9603. 2018. View Article : Google Scholar : PubMed/NCBI

66 

Sheikh SR, Patel NJ and Recinos VMR: Safety and technical efficacy of pediatric brainstem biopsies: An updated meta-analysis of 1000+ children. World Neurosurg. 189:428–438.e2. 2024. View Article : Google Scholar : PubMed/NCBI

67 

Walker DA, Liu J, Kieran M, Jabado N, Picton S, Packer R and St Rose C; CPN Paris 2011 Conference Consensus Group, : A multi-disciplinary consensus statement concerning surgical approaches to Low-grade, High-grade astrocytomas and diffuse intrinsic pontine gliomas in childhood (CPN Paris 2011) using the Delphi method. Neuro Oncol. 15:462–468. 2013. View Article : Google Scholar : PubMed/NCBI

68 

Ryba A, Özdemir Z, Nissimov N, Hönikl L, Neidert N, Jakobs M, Kalasauskas D, Krigers A, Thomé C, Freyschlag CF, et al: 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. 26:1479–1493. 2024. View Article : Google Scholar : PubMed/NCBI

69 

Dalmage M, LoPresti MA, Sarkar P, Ranganathan S, Abdelmageed S, Pagadala M, Shlobin NA, Lam S and DeCuypere M: Survival and neurological outcomes after stereotactic biopsy of diffuse intrinsic pontine glioma: A systematic review. J Neurosurg Pediatr. 32:665–672. 2023. View Article : Google Scholar : PubMed/NCBI

70 

Gajjar A, Mahajan A, Abdelbaki M, Anderson C, Antony R, Bale T, Bindra R, Bowers DC, Cohen K, Cole B, et al: Pediatric central nervous system cancers, version 2.2023, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 20:1339–1362. 2022.PubMed/NCBI

71 

Kim HJ and Suh CO: Radiotherapy for diffuse intrinsic pontine glioma: Insufficient but indispensable. Brain Tumor Res Treat. 11:79–85. 2023. View Article : Google Scholar : PubMed/NCBI

72 

Cohen KJ, Jabado N and Grill J: Diffuse intrinsic pontine gliomas-current management and new biologic insights. Is there a glimmer of hope? Neuro Oncol. 19:1025–1034. 2017.PubMed/NCBI

73 

Wawrzuta D, Chojnacka M, Drogosiewicz M, Pedziwiatr K and Dembowska-Baginska B: Reirradiation for diffuse intrinsic pontine glioma: Prognostic radiomic factors at progression. Strahlenther Onkol. 200:797–804. 2024. View Article : Google Scholar : PubMed/NCBI

74 

Esparragosa Vazquez I and Ducray F: The role of radiotherapy, chemotherapy, and targeted therapies in adult intramedullary spinal cord tumors. Cancers (Basel). 16:27812024. View Article : Google Scholar : PubMed/NCBI

75 

Yang Z, Sun L, Chen H, Sun C and Xia L: New progress in the treatment of diffuse midline glioma with H3K27M alteration. Heliyon. 10:e248772024. View Article : Google Scholar : PubMed/NCBI

76 

Argersinger DP, Rivas SR, Shah AH, Jackson S and Heiss JD: New developments in the pathogenesis, therapeutic targeting, and treatment of H3K27M-mutant diffuse midline glioma. Cancers (Basel). 13:52802021. View Article : Google Scholar : PubMed/NCBI

77 

Mandorino M, Maitra A, Armenise D, Baldelli OM, Miciaccia M, Ferorelli S, Perrone MG and Scilimati A: Pediatric diffuse midline glioma H3K27-altered: From developmental origins to therapeutic challenges. Cancers (Basel). 16:18142024. View Article : Google Scholar : PubMed/NCBI

78 

Watanabe J, Clutter MR, Gullette MJ, Sasaki T, Uchida E, Kaur S, Mo Y, Abe K, Ishi Y, Takata N, et al: BET bromodomain inhibition potentiates radiosensitivity in models of H3K27-altered diffuse midline glioma. J Clin Invest. 134:e1747942024. View Article : Google Scholar : PubMed/NCBI

79 

Bailey S, Howman A, Wheatley K, Wherton D, Boota N, Pizer B, Fisher D, Kearns P, Picton S, Saran F, et al: Diffuse intrinsic pontine glioma treated with prolonged temozolomide and radiotherapy-results of a United Kingdom phase II trial (CNS 2007 04). Eur J Cancer. 49:3856–3862. 2013. View Article : Google Scholar : PubMed/NCBI

80 

Chassot A, Canale S, Varlet P, Puget S, Roujeau T, Negretti L, Dhermain F, Rialland X, Raquin MA, Grill J, et al: Radiotherapy with concurrent and adjuvant temozolomide in children with newly diagnosed diffuse intrinsic pontine glioma. J Neurooncol. 106:399–407. 2012. View Article : Google Scholar : PubMed/NCBI

81 

Abe H, Natsumeda M, Kanemaru Y, Watanabe J, Tsukamoto Y, Okada M, Yoshimura J, Oishi M and Fujii Y: MGMT expression contributes to temozolomide resistance in H3K27M-mutant diffuse midline gliomas and MGMT silencing to temozolomide sensitivity in IDH-mutant gliomas. Neurol Med Chir (Tokyo). 58:290–295. 2018. View Article : Google Scholar : PubMed/NCBI

82 

Brandes AA, Tosoni A, Cavallo G, Reni M, Franceschi E, Bonaldi L, Bertorelle R, Gardiman M, Ghimenton C, Iuzzolino P, et al: Correlations between 06-methylguanine DNA methyltransferase promoter methylation status, 1p and 19q deletions, and response to temozolomide in anaplastic and recurrent oligodendroglioma: A prospective GICNO study. J Clin Oncol. 24:4746–4753. 2006. View Article : Google Scholar : PubMed/NCBI

83 

Warren KE, Aikin AA, Libucha M, Widemann BC, Fox E, Packer RJ and Balis FM: Phase I study of O6-benzylguanine and temozolomide administered daily for 5 days to pediatric patients with solid tumors. J Clin Oncol. 23:7646–7653. 2005. View Article : Google Scholar : PubMed/NCBI

84 

Zhang Y, Dong W, Zhu J, Wang L, Wu X and Shan H: Combination of EZH2 inhibitor and BET inhibitor for treatment of diffuse intrinsic pontine glioma. Cell Biosci. 7:562017. View Article : Google Scholar : PubMed/NCBI

85 

Sasaki T, Katagi H, Goldman S, Becher OJ and Hashizume R: Convection-enhanced delivery of enhancer of zeste homolog-2 (EZH2) inhibitor for the treatment of diffuse intrinsic pontine glioma. Neurosurgery. 87:E680–E688. 2020. View Article : Google Scholar : PubMed/NCBI

86 

Singleton WGB, Bienemann AS, Woolley M, Johnson D, Lewis O, Wyatt MJ, Damment SJP, Boulter LJ, Killick-Cole CL, Asby DJ and Gill SS: The distribution, clearance, and brainstem toxicity of panobinostat administered by convection-enhanced delivery. J Neurosurg Pediatr. 22:288–296. 2018. View Article : Google Scholar : PubMed/NCBI

87 

Massimino M, Spreafico F, Biassoni V, Simonetti F, Riva D, Trecate G, Giombini S, Poggi G, Pecori E, Pignoli E, et al: Diffuse pontine gliomas in children: Changing strategies, changing results? A mono-institutional 20-year experience. J Neurooncol. 87:355–361. 2008. View Article : Google Scholar : PubMed/NCBI

88 

Tsoli M, Ung C, Upton DH, Venkat P, Salerno A, Pandher R, Mayoh C, Vittorio O and Ziegler DS: Dipg-35. Overcoming the blood-brain barrier challenge in diffuse midline glioma. Neuro Oncol. 26 (Suppl 4):02024. View Article : Google Scholar

89 

Ono T, Kuwashige H, Adachi JI, Takahashi M, Oda M, Kumabe T and Shimizu H: Long-term survival of a patient with diffuse midline glioma in the pineal region: A case report and literature review. Surg Neurol Int. 12:6122021. View Article : Google Scholar : PubMed/NCBI

90 

Veldhuijzen van Zanten SEM, El-Khouly FE, Jansen MHA, Bakker DP, Sanchez Aliaga E, Haasbeek CJA, Wolf NI, Zwaan CM, Vandertop WP, van Vuurden DG and Kaspers GJL: A phase I/II study of gemcitabine during radiotherapy in children with newly diagnosed diffuse intrinsic pontine glioma. J Neurooncol. 135:307–315. 2017. View Article : Google Scholar : PubMed/NCBI

91 

Kilburn LB, Kocak M, Baxter P, Poussaint TY, Paulino AC, McIntyre C, Lemenuel-Diot A, Lopez-Diaz C, Kun L, Chintagumpala M, et al: A pediatric brain tumor consortium phase II trial of capecitabine rapidly disintegrating tablets with concomitant radiation therapy in children with newly diagnosed diffuse intrinsic pontine gliomas. Pediatr Blood Cancer. 65:10.1002/pbc.26832. 2018. View Article : Google Scholar

92 

Zhang S, Yang X, Tan Q, Sun H, Chen D, Chen Y, Zhang H, Yang Y, Gong Q and Yue Q: Cortical myelin and thickness mapping provide insights into whole-brain tumor burden in diffuse midline glioma. Cereb Cortex. 34:bhad4912024. View Article : Google Scholar : PubMed/NCBI

93 

Krystal J, Hanson D, Donnelly D and Atlas M: A phase 1 study of mebendazole with bevacizumab and irinotecan in high-grade gliomas. Pediatr Blood Cancer. 71:e308742024. View Article : Google Scholar : PubMed/NCBI

94 

Voelcker G: The mechanism of action of cyclophosphamide and its consequences for the development of a new generation of oxazaphosphorine cytostatics. Sci Pharm. 88:422020. View Article : Google Scholar

95 

Lin FY, Stuckert A, Tat C, White M, Ruggieri L, Zhang H, Mehta B, Lapteva N, Mei Z, Major A, et al: Phase I trial of GD2.CART cells augmented with constitutive interleukin-7 receptor for treatment of high-grade pediatric CNS tumors. J Clin Oncol. 42:2769–2779. 2024. View Article : Google Scholar : PubMed/NCBI

96 

Ishizawa J, Zarabi SF, Davis RE, Halgas O, Nii T, Jitkova Y, Zhao R, St-Germain J, Heese LE, Egan G, et al: Mitochondrial ClpP-mediated proteolysis induces selective cancer cell lethality. Cancer Cell. 35:721–737.e9. 2019. View Article : Google Scholar : PubMed/NCBI

97 

Jacques S, van der Sloot AM, C Huard C, Coulombe-Huntington J, Tsao S, Tollis S, Bertomeu T, Culp EJ, Pallant D, Cook MA, et al: Imipridone anticancer compounds ectopically activate the ClpP protease and represent a new scaffold for antibiotic development. Genetics. 214:1103–1120. 2020. View Article : Google Scholar : PubMed/NCBI

98 

Kline CL, Van den Heuvel AP, Allen JE, Prabhu VV, Dicker DT and El-Deiry WS: ONC201 kills solid tumor cells by triggering an integrated stress response dependent on ATF4 activation by specific eIF2alpha kinases. Sci Signal. 9:ra182016. View Article : Google Scholar : PubMed/NCBI

99 

Prabhu VV, Morrow S, Rahman Kawakibi A, Zhou L, Ralff M, Ray J, Jhaveri A, Ferrarini I, Lee Y, Parker C, et al: ONC201 and imipridones: Anti-cancer compounds with clinical efficacy. Neoplasia. 22:725–744. 2020. View Article : Google Scholar : PubMed/NCBI

100 

Venneti S, Kawakibi AR, Ji S, Waszak SM, Sweha SR, Mota M, Pun M, Deogharkar A, Chung C, Tarapore RS, et al: Clinical efficacy of ONC201 in H3K27M-mutant diffuse midline gliomas is driven by disruption of integrated metabolic and epigenetic pathways. Cancer Discov. 13:2370–2393. 2023. View Article : Google Scholar : PubMed/NCBI

101 

Arrillaga-Romany I, Lassman A, McGovern SL, Mueller S, Nabors B, van den Bent M, Vogelbaum MA, Allen JE, Melemed AS, Tarapore RS, et al: ACTION: A randomized phase 3 study of ONC201 (dordaviprone) in patients with newly diagnosed H3 K27M-mutant diffuse glioma. Neuro Oncol. 26 (Suppl_2):S173–S181. 2024. View Article : Google Scholar : PubMed/NCBI

102 

Jenke R, Ressing N, Hansen FK, Aigner A and Buch T: Anticancer therapy with HDAC inhibitors: Mechanism-based combination strategies and future perspectives. Cancers (Basel). 13:6342021. View Article : Google Scholar : PubMed/NCBI

103 

Eckschlager T, Plch J, Stiborova M and Hrabeta J: Histone deacetylase inhibitors as anticancer drugs. Int J Mol Sci. 18:14142017. View Article : Google Scholar : PubMed/NCBI

104 

Mueller S, Kline C, Stoller S, Lundy S, Christopher L, Reddy AT, Banerjee A, Cooney TM, Raber S, Hoffman C, et al: PNOC015: Repeated convection-enhanced delivery of MTX110 (aqueous panobinostat) in children with newly diagnosed diffuse intrinsic pontine glioma. Neuro Oncol. 25:2074–2086. 2023. View Article : Google Scholar : PubMed/NCBI

105 

Su JM, Kilburn LB, Mansur DB, Krailo M, Buxton A, Adekunle A, Gajjar A, Adamson PC, Weigel B, Fox E, et al: Phase I/II trial of vorinostat and radiation and maintenance vorinostat in children with diffuse intrinsic pontine glioma: A Children's Oncology Group report. Neuro Oncol. 24:655–664. 2022. View Article : Google Scholar : PubMed/NCBI

106 

Neth BJ, Balakrishnan SN, Carabenciov ID, Uhm JH, Daniels DJ, Kizilbash SH and Ruff MW: Panobinostat in adults with H3 K27M-mutant diffuse midline glioma: A single-center experience. J Neurooncol. 157:91–100. 2022. View Article : Google Scholar : PubMed/NCBI

107 

Ameratunga M, Kipps E, Okines AFC and Lopez JS: To cycle or Fight-CDK4/6 inhibitors at the crossroads of anticancer immunity. Clin Cancer Res. 25:21–28. 2019. View Article : Google Scholar : PubMed/NCBI

108 

Zhang J, Xu D, Zhou Y, Zhu Z and Yang X: Mechanisms and implications of CDK4/6 inhibitors for the treatment of NSCLC. Front Oncol. 11:6760412021. View Article : Google Scholar : PubMed/NCBI

109 

The JLF and Aplin AE: Arrested developments: CDK4/6 inhibitor resistance and alterations in the tumor immune microenvironment. Clin Cancer Res. 25:921–927. 2019. View Article : Google Scholar : PubMed/NCBI

110 

Previtali R, Prontera G, Alfei E, Nespoli L, Masnada S, Veggiotti P and Mannarino S: Paradigm shift in the treatment of tuberous sclerosis: Effectiveness of everolimus. Pharmacol Res. 195:1068842023. View Article : Google Scholar : PubMed/NCBI

111 

DeWire M, Fuller C, Hummel TR, Chow LML, Salloum R, de Blank P, Pater L, Lawson S, Zhu X, Dexheimer P, et al: A phase I/II study of ribociclib following radiation therapy in children with newly diagnosed diffuse intrinsic pontine glioma (DIPG). J Neurooncol. 149:511–522. 2020. View Article : Google Scholar : PubMed/NCBI

112 

DeWire M, Lazow M, Campagne O, Leach J, Fuller C, Senthil Kumar S, Stanek J, de Blank P, Hummel TR, Pillay-Smiley N, et al: Phase I study of ribociclib and everolimus in children with newly diagnosed DIPG and high-grade glioma: A CONNECT pediatric neuro-oncology consortium report. Neurooncol Adv. 4:vdac0552022.PubMed/NCBI

113 

Greenall SA, McKenzie M, Seminova E, Dolezal O, Pearce L, Bentley J, Kuchibhotla M, Chen SC, McDonald KL, Kornblum HI, et al: Most clinical anti-EGFR antibodies do not neutralize both wtEGFR and EGFRvIII activation in glioma. Neuro Oncol. 21:1016–1027. 2019. View Article : Google Scholar : PubMed/NCBI

114 

Fleischhack G, Massimino M, Warmuth-Metz M, Khuhlaeva E, Janssen G, Graf N, Rutkowski S, Beilken A, Schmid I, Biassoni V, et al: Nimotuzumab and radiotherapy for treatment of newly diagnosed diffuse intrinsic pontine glioma (DIPG): A phase III clinical study. J Neurooncol. 143:107–113. 2019. View Article : Google Scholar : PubMed/NCBI

115 

Li S, Wang C, Chen J, Lan Y, Zhang W, Kang Z, Zheng Y, Zhang R, Yu J and Li W: Signaling pathways in brain tumors and therapeutic interventions. Signal Transduct Target Ther. 8:82023. View Article : Google Scholar : PubMed/NCBI

116 

Meel MH, Kaspers GJL and Hulleman E: Preclinical therapeutic targets in diffuse midline glioma. Drug Resist Updat. 44:15–25. 2019. View Article : Google Scholar : PubMed/NCBI

117 

Yang K, Wu Z, Zhang H, Zhang N, Wu W, Wang Z, Dai Z, Zhang X, Zhang L, Peng Y, et al: Glioma targeted therapy: Insight into future of molecular approaches. Mol Cancer. 21:392022. View Article : Google Scholar : PubMed/NCBI

118 

Liu Y, Liu C, Wang G, Tao R, Xu J, Shen L, Wang H, Zhu C, Zhang H, Zeng G, et al: Nimotuzumab combined with chemoradiation therapy in newly diagnosed pediatric diffuse intrinsic pontine glioma. Int J Radiat Oncol Biol Phys. 123:839–847. 2025. View Article : Google Scholar : PubMed/NCBI

119 

Pachocki CJ and Hol EM: Current perspectives on diffuse midline glioma and a different role for the immune microenvironment compared to glioblastoma. J Neuroinflammation. 19:2762022. View Article : Google Scholar : PubMed/NCBI

120 

Majzner RG, Ramakrishna S, Yeom KW, Patel S, Chinnasamy H, Schultz LM, Richards RM, Jiang L, Barsan V, Mancusi R, et al: GD2-CART cell therapy for H3K27M-mutated diffuse midline gliomas. Nature. 603:934–941. 2022. View Article : Google Scholar : PubMed/NCBI

121 

Vitanza NA, Wilson AL, Huang W, Seidel K, Brown C, Gustafson JA, Yokoyama JK, Johnson AJ, Baxter BA, Koning RW, et al: Intraventricular B7-H3 CAR T cells for diffuse intrinsic pontine glioma: Preliminary First-in-Human bioactivity and safety. Cancer Discov. 13:114–131. 2023. View Article : Google Scholar : PubMed/NCBI

122 

Vitanza NA, Ronsley R, Choe M, Seidel K, Huang W, Rawlings-Rhea SD, Beam M, Steinmetzer L, Wilson AL, Brown C, et al: Intracerebroventricular B7-H3-targeting CAR T cells for diffuse intrinsic pontine glioma: A phase 1 trial. Nat Med. 31:861–868. 2025. View Article : Google Scholar : PubMed/NCBI

123 

Dai B, Roife D, Kang Y, Gumin J, Rios Perez MV, Li X, Pratt M, Brekken RA, Fueyo-Margareto J, Lang FF, et al: Preclinical evaluation of sequential combination of oncolytic adenovirus delta-24-RGD and Phosphatidylserine-targeting antibody in pancreatic ductal adenocarcinoma. Mol Cancer Ther. 16:662–670. 2017. View Article : Google Scholar : PubMed/NCBI

124 

Gállego Pérez-Larraya J, Garcia-Moure M, Labiano S, Patiño-García A, Dobbs J, Gonzalez-Huarriz M, Zalacain M, Marrodan L, Martinez-Velez N, Puigdelloses M, et al: Oncolytic DNX-2401 virus for pediatric diffuse intrinsic pontine glioma. N Engl J Med. 386:2471–2481. 2022. View Article : Google Scholar : PubMed/NCBI

125 

Grassl N, Poschke I, Lindner K, Bunse L, Mildenberger I, Boschert T, Jähne K, Green EW, Hülsmeyer I, Jünger S, et al: A H3K27M-targeted vaccine in adults with diffuse midline glioma. Nat Med. 29:2586–2592. 2023. View Article : Google Scholar : PubMed/NCBI

126 

Boschert T, Kromer K, Lerner T, Lindner K, Haltenhof G, Tan CL, Jähne K, Poschke I, Bunse L, Eisele P, et al: H3K27M neoepitope vaccination in diffuse midline glioma induces B and T cell responses across diverse HLA loci of a recovered patient. Sci Adv. 10:eadi90912024. View Article : Google Scholar : PubMed/NCBI

127 

Vincent CA and Remeseiro S: The immune response behind peptide vaccination in diffuse midline glioma. Mol Oncol. 18:1849–1852. 2024. View Article : Google Scholar : PubMed/NCBI

128 

Mueller S, Taitt JM, Villanueva-Meyer JE, Bonner ER, Nejo T, Lulla RR, Goldman S, Banerjee A, Chi SN, Whipple NS, et al: Mass cytometry detects H3.3K27M-specific vaccine responses in diffuse midline glioma. J Clin Investigation. 130:6325–6337. 2020. View Article : Google Scholar : PubMed/NCBI

129 

Grassl N, Sahm K, Süße H, Poschke I, Bunse L, Bunse T, Boschert T, Mildenberger I, Rupp AK, Ewinger MP, et al: INTERCEPT H3: A multicenter phase I peptide vaccine trial for the treatment of H3-mutated diffuse midline gliomas. Neurol Res Pract. 5:552023. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Wu W, Chen W, Ma W and Wang Y: <p>Epigenetic mechanisms and therapeutic advances in diffuse midline glioma (Review)</p>. Oncol Rep 55: 50, 2026.
APA
Wu, W., Chen, W., Ma, W., & Wang, Y. (2026). <p>Epigenetic mechanisms and therapeutic advances in diffuse midline glioma (Review)</p>. Oncology Reports, 55, 50. https://doi.org/10.3892/or.2026.9055
MLA
Wu, W., Chen, W., Ma, W., Wang, Y."<p>Epigenetic mechanisms and therapeutic advances in diffuse midline glioma (Review)</p>". Oncology Reports 55.3 (2026): 50.
Chicago
Wu, W., Chen, W., Ma, W., Wang, Y."<p>Epigenetic mechanisms and therapeutic advances in diffuse midline glioma (Review)</p>". Oncology Reports 55, no. 3 (2026): 50. https://doi.org/10.3892/or.2026.9055
Copy and paste a formatted citation
x
Spandidos Publications style
Wu W, Chen W, Ma W and Wang Y: <p>Epigenetic mechanisms and therapeutic advances in diffuse midline glioma (Review)</p>. Oncol Rep 55: 50, 2026.
APA
Wu, W., Chen, W., Ma, W., & Wang, Y. (2026). <p>Epigenetic mechanisms and therapeutic advances in diffuse midline glioma (Review)</p>. Oncology Reports, 55, 50. https://doi.org/10.3892/or.2026.9055
MLA
Wu, W., Chen, W., Ma, W., Wang, Y."<p>Epigenetic mechanisms and therapeutic advances in diffuse midline glioma (Review)</p>". Oncology Reports 55.3 (2026): 50.
Chicago
Wu, W., Chen, W., Ma, W., Wang, Y."<p>Epigenetic mechanisms and therapeutic advances in diffuse midline glioma (Review)</p>". Oncology Reports 55, no. 3 (2026): 50. https://doi.org/10.3892/or.2026.9055
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team