Next Article in Journal
Mechanism of Sepsis
Previous Article in Journal
Methylene Blue Increases Active Mitochondria and Cellular Survival Through Modulation of miR16–UPR Signaling Axis
Previous Article in Special Issue
The Current Landscape of Molecular Pathology for the Diagnosis and Treatment of Atypical Teratoid Rhabdoid Tumor
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Current Landscape of Molecular Pathology for the Diagnosis and Treatment of Pediatric High-Grade Glioma

1
Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229-3039, USA
2
Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
3
Cancer and Blood Diseases Institute, The Cure Starts Now Foundation Brain Tumor Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229-3039, USA
*
Author to whom correspondence should be addressed.
J. Mol. Pathol. 2025, 6(3), 17; https://doi.org/10.3390/jmp6030017
Submission received: 23 April 2025 / Revised: 8 July 2025 / Accepted: 29 July 2025 / Published: 1 August 2025
(This article belongs to the Collection Feature Papers in Journal of Molecular Pathology)

Abstract

Pediatric high-grade glioma (pHGG) is a devastating group of childhood cancers associated with poor outcomes. Traditionally, diagnosis was based on histologic and immunohistochemical characteristics, including high mitotic activity, presence of necrosis, and presence of glial cell markers (e.g., GFAP). With advances in molecular tumor profiling, these tumors have been recategorized based on specific molecular findings that better lend themselves to prediction of treatment response and prognosis. pHGG is now categorized into four subtypes: H3K27-altered, H3G34-mutant, H3/IDH-WT, and infant-type high-grade glioma (iHGG). Molecular profiling has not only increased the specificity of diagnosis but also improved prognostication. Additionally, these molecular findings provide novel targets for individual tumor-directed therapy. While these therapies are largely still under investigation, continued investigation of distinct molecular markers in these tumors is imperative to extending event-free survival (EFS) and overall survival (OS) for patients with pHGG.

1. Introduction

Pediatric high-grade gliomas (pHGG) are a group of highly aggressive tumors and are the leading cause of cancer-related death in children [1]. It has only been in the last two decades that pHGG has been recognized as a distinct, biologically unique entity from adult gliomas [2]. Until recent years, pathologists and oncologists relied on histologic and immunohistochemical findings in combination with clinical symptomology and tumor location to diagnose patients with a pHGG that was distinct from a pediatric low-grade glioma (pLGG). Despite the ability to generalize tumors based on these findings, pHGG and pLGG frequently have overlapping histopathologic findings making it difficult to predict response to treatment and clinical outcomes. Characteristic imaging findings have also historically played a role in diagnosis and differentiation, with pHGGs primarily appearing as heterogenous masses on MRI with or without enhancement, in comparison to the well-circumscribed lesions seen in pLGG; however, imaging alone is not adequate for distinguishing between types of pHGG [3]. It is only with advances in genetic and molecular profiling of pHGG that more specific diagnoses have become widely accepted. pHGG is now grouped into four subtypes as defined by the WHO CNS5 2021 guidelines with specific associated molecular and genetic changes (Table 1). Additionally, pHGG has historically been treated with surgical resection followed by radiation and/or chemotherapy, typically temozolomide, as this agent has been proven effective in adult high-grade glioma (aHGG) [4]. Further profiling has expanded the current knowledge of oncogenesis and provides new avenues for treatment of these devastating tumors that align with the knowledge that pHGG is its own unique entity from aHGG [5]. This review describes the molecular subtypes of pHGG and how an understanding of these molecular differences allows for identification of novel targets for therapy.

2. pHGG Diagnoses and Therapeutic Targets

2.1. Diffuse Midline Glioma (DMG), H3K27-Altered

Perhaps one of the best examples of the need for molecular profiling in the classification and risk stratification of pHGG is seen with diffuse midline gliomas (DMG). Histologically, this tumor type is characterized by hypercellular, diffuse astrocytic morphology with high mitotic activity and varying amounts of necrosis [6]. The immunophenotype is consistent with its glial cell origin, with positive staining for GFAP and OLIG2. Historically, the degree of mitotic activity and necrotic tissue was used to diagnose and risk-stratify these tumors. Molecular profiling has shown that certain mutations place this tumor type in a high-risk category (CNS WHO grade 4) despite histologic findings [7]. The most well-known tumor in this group is the diffuse intrinsic pontine glioma (DIPG). DIPGs were not historically biopsied and were instead diagnosed based on classic MRI findings of at least 50% involvement of the pons associated with T1-hypointensity and T2-hyperintensity, so it was not possible to molecularly understand these tumors [8]. In recent decades as biopsy has become more common and safer, a specific histone mutation was found to be commonly associated with DIPGs in addition to other midline HGGs such as primary spinal cord or thalamic tumors, thus the DMG group was formed. This emphasizes the importance of molecular findings when diagnosing pHGGs.
Subtypes of DMG include DMG H3K27-altered, EGFR-mutant, and H3-WT with EZHIP overexpression [9]. The most common of these is DMG H3K27-altered, which is most commonly seen in young adults with studies reporting a median age of 14 [6]. These tumors are exclusively found along the midline and have a wide variety of histologic appearances. The majority exhibit hypercellularity and have an astrocytoma-like morphology with extensive mitotic activity and varying degrees of necrosis [6]. Given its histologic diversity, further molecular characterization is beneficial for diagnosis. The H3K27-altered group of tumors is a histone mutation-driven tumor, the first of which were identified in pHGG [1]. It is the result of missense mutations on the H3F3A gene affecting either histone 3.3 (DMG H3.3K27) or histone 3.1 (DMG H3.1K27) leading to replacement of lysine with methionine [10]. This results in downregulation of H3K27me3 and resultant amplification of platelet-derived growth factor (PDGFRA) and ACVR1 mutations, respectively [1]. These mutations typically co-occur with mutations in either tumor suppressor protein 53 (TP53) or PIK3R1, resulting in additional impacts to cell-cycle regulation and growth factor signaling [11]. H3K27-altered tumors have been shown to be associated with generally poorer prognosis in comparison to wild-type variants of pHGG, with a reported 99% 5-year mortality rate and median survival of approximately 11 months [6,10]. This is perhaps due to its low level of MGMT promoter methylation, which is associated with poorer response to traditional chemotherapy treatment with temozolomide [4]. However, the subset of these with ACVR1 mutations have exhibited slightly longer survival than other subsets [12]. Identification of these mutations and molecular markers also provides multiple avenues for targeted therapies.
Several clinical trials have been completed while others are actively enrolling to identify appropriate therapeutic targets for DMG, H3K27-altered tumors. The current standard of care is radiation therapy which prolongs progression-free survival without significant effects on overall survival (OS). In most pediatric oncology centers, an early phase clinical trial is considered as upfront therapy either concurrent with or post radiation. The multitude of trials either target molecularly altered pathways, genetic alterations in the tumor, or attempt to harness the immune environment as a mode of therapy [13]. For example, histone deacetylase (HDAC) is an enzyme involved in the removal of acetyl groups from histone tails. HDAC inhibitors such as Panobinostat have been shown to restore histone 3 (H3) acetylation, reducing cellular proliferation; however, clinical effectiveness has been suboptimal most likely due to poor penetration through the blood–brain barrier [14,15]. For the subset of DMG H3K27-altered tumors that overexpress PDGFRA, tyrosine kinase inhibitors have been shown to be effective at inhibiting PDGFRA and may be useful as a therapeutic option [14,16]. Other trials are also focusing on dual target therapy, such as CUDC-907, a HDAC and PI3K inhibitor, for the treatment of H3K27-altered DMG [14]. There are also several ongoing immunotherapeutic trials including the use of CAR-T cells such as those at Stanford and at Seattle Children’s [17,18].

2.2. Diffuse Hemispheric Glioma (DHG), H3G34-Mutant

The H3G34-mutant pHGGs are most commonly seen in young adults, with one study noting a median age of 19 [19]. These tumors typically arise in supratentorial spaces—most often the temporal and parietal lobes [10,19]. Similar to DMG H3K27-altered tumors, the DHG H3G34-mutant tumors are typically characterized histologically by high-grade astrocytic morphology. A subset of these have an embryonal-like appearance with a high nucleus to cytoplasm ratio, with associated vascular proliferation and necrosis [6,7]. Like the H3K27-altered tumors, GFAP is expressed with immunohistochemical staining, though conversely OLIG2 is not often expressed in these tumors [10]. Given their histopathologic and immunohistochemical similarities, the H3G34-mutant was not identified as a separate entity from the H3K27-altered tumors until much more recently following identification by molecular profiling [10]. Based on these findings alone, H3G34-mutant tumors were previously diagnosed as pHGG or as a primitive neuroectodermal tumor, resulting in minimally effective treatment selection and poor ability to predict outcomes [20]. Current treatment of these tumors primarily consists of surgical resection, followed by radiation therapy and chemotherapy, though a standardized chemotherapeutic regimen for pediatric-type H3G34-mutants does not currently exist [20]. Median OS is most often reported to be between 14 and 17 months, which is slightly improved from that of the H3K27-altered tumors; this may be secondary to ease of surgical resection based on accessibility of hemispheric versus midline gliomas [10,20].
The DHG H3G34-mutant is another example of a histone mutation-driven tumor, with its defining mutation on the H3-3A gene with resultant replacement of glycine with arginine or glycine with valine (H3G34R/V), with H3G34R the most common mutation found in DHG H3G34-mutants [10,20]. This replacement results in decreased SETD2 methyltransferase activity, leading to decreased levels of H3K36me3 [7,20]. Decreased H3K36me3 has been shown to be involved in the methylation of H3K27, resulting in increased levels of H3K27me3, in contrast to the downregulation of this post-translational modification seen in H3K27-altered gliomas [1,20]. These tumors are frequently found to have methylation of the MGMT promoter, despite global DNA hypomethylation resulting from decreased interaction with and redistribution of DNA methyltransferase 1 noted in these tumors [6,7,20]. These molecular findings can be beneficial in risk stratification; for example, tumors with MGMT promoter methylation have been shown to have a better prognosis than those characterized by amplification of oncogenes (e.g., EGFR, MDM2) [7]. Decreased H3K36me3 has also been implicated in defects in DNA mismatch repair (MMR), possibly contributing to tumorigenesis [20]. H3G34-mutants are additionally associated with mutations in TP53, a tumor suppressor protein, and ATRX, a protein involved in chromatin remodeling important for genetic stability; PDGFRA amplification is also commonly found [6]. These tumors are also associated with amplification of MYCN [6].
Identified molecular changes allow for various treatment targets. One of these targets is Aurora Kinase A (AURKA), a regulator of MYCN; AURKA inhibitors have shown selectivity against cells of DHG H3G34-mutant tumors [21]. The PDGFRA pathway has also been identified as a therapeutic target in these tumors; PDGFRA inhibitors in combination with MET inhibitors or mTOR inhibitors showed improved OS in vivo [22]. Cell-cycle components have also been noted to be more frequently found in H3G34-mutants, opening the possibility for cyclin-dependent kinase inhibitors as potential targets for treatment [10]. There are few clinical trials focusing on molecular targets of DHG H3G34-mutant tumors when compared to those for the DMG H3K27-altered tumors, requiring further investigation [10].

2.3. Diffuse Pediatric Type HGG, H3-Wild Type and IDH-Wild Type (pHGG H3/IDH-WT)

Akin to the H3G34-mutant, the pHGG H3/IDH-WT subgroup of pHGG was recently identified as a distinct tumor class. Its definition is reliant upon the lack of mutations in both IDH, common in pHGG, and the histone mutations found in the DMG H3K27-altered and DHG H3G34-mutants [23]. These tumors are histologically diverse. Hong et al. showed these tumors to be associated with TP53 and ATRX mutations, as well as mutations of MMR genes, with those harboring TP53 mutations most specifically associated with adverse outcomes [23]. Given the presence of MMR genes, these tumors have also been shown to be associated with familial cancer syndromes such as Lynch syndrome and Li-Fraumeni [9,23]. These tumors have been subcategorized into the following methylation profiles: RTK1, RTK2, and MYCN. The MYCN methylation subtype is characterized by resultant amplification of MYCN and often the amplification of ID2 [24]. These tumors are the most common subtype associated with TP53 mutations, as well as pHGG occurring in the context of Li Fraumeni [6,24]. In comparison to other subtypes, these tumors may be more likely located infratentorially, particularly in the brain stem [24]. Tumors with the RTK1 methylation profiles have been shown to be located either supratentorially or infratentorially, whereas the RTK2 profile tumors are most often supratentorial [6,24]. These subtypes are associated with amplification of PDGFRA and EGFR, respectively, resulting in disturbances in cellular signaling leading to oncogenesis; the RTK1 profile is most likely to represent those with pHGG in the context of Lynch syndrome [6,25]. The RTK2 methylation profile has also been shown to have a higher frequency of TERT-promoter mutations, and is associated with highest OS rate in comparison to other pHGG H3/IDH-WT tumors, with a 5-year OS of approximately 50% [24]. Other studies have not found a correlation between methylation profiles and their associated amplifications and OS [23,24,26]. The MYCN subgroup OS has been shown to be closer to that of the H3K27-altered pHGG with median OS of 17.2 months; this may be secondary to the more frequent association of this methylation profile with mutations in TP53 [24,26].
Though advances in molecular diagnostics have allowed for further characterization of these tumors, PDGFRA, EGFR, and/or MYCN amplification are nonspecific findings and have been seen in other subcategories of pHGG. Additionally, tumors have been identified as not having IDH or H3 mutations and do not fall into any of the methylation profiles previously described [7]. Further research into H3/IDH-WT tumors is necessary for accurate subcategorization and risk stratification.

2.4. Infant Type Hemispheric Glioma (iHGG)

While the exact definition of what constitutes an infantile brain tumor remains up for debate, most agree that these are tumors occur within the first year of life. iHGG, histologically, is typically described as having astrocytic-like cells, as well as occasional spindle cells [9]. Advances in molecular profiling have allowed them to be differentiated from other neoplasms of infancy, and other histologically similar tumors [27]. Alterations in RAS/MAPK seen in tumors in this age group are most often categorized as pLGG, whereas those containing mutations in RTK oncogenes are considered pHGG. The patients are therefore diagnosed with iHGG despite the latter group often exhibiting methylation profiles similar to pLGGs [28,29]. These tumors are histologically characterized by sheets of astrocytic-type cells and focal spindled cells with a definitive separation between neoplastic and non-neoplastic tissue [12,27].
iHGG is currently defined by the presence of receptor tyrosine kinase (RTK) gene fusion, with reports of over 70% of iHGG containing these fusions [30]. These tumors can be molecularly subdivided based on type of RTK gene fusion: NTRK-altered, ALK-altered, ROS-altered, and MET-altered [7,9]. While these fusions may be associated with both pLGG and pHGG, over 80% (24/29) of tumors harboring these fusions have been shown to be enriched for pHGG [28]. Clarke et al. described an “intrinsic set” of infant gliomas (n = 130) based on fusion gene analysis and methylation profiling, 21 of which were considered iHGG. Of these, 10/21 (48%) were noted to contain ALK-fusions, 12/21 (57%) were noted to have NTRK-fusions, with ROS- and MET-fusions representing 9% (2/21) and 5% (1/21), respectively [29]. Smaller studies have confirmed the higher frequency of these mutations, reporting 27% (3/11) of an iHGG cohort containing NTRK-fusions [31]. Numerous chromosomal rearrangements are also noted in iHGGs. Most are associated with dysregulation of genes involved in development, including WNT signaling pathways. Molecular profiling by next generation sequencing has also contributed to better predication of OS, with the subset of iHGG containing chromosomal fusions having increased OS in comparison to the smaller subgroup without fusions [29]. The methylation profile of these tumors has been shown to be more akin to that of pLGGs which may also related to improved OS [27,32].
Due to RTKs being heavily implicated in tumorigenesis of iHGG, RTK inhibitors have been shown to have promising anti-tumor activity in pediatric patients [27]. ALK inhibitors have specifically shown to be an effective target of treatment amongst this subgroup of pHGG; for example, a combined ALK and ROS1 inhibitor has been developed as viable treatment option for these tumors, though clinical trials are ongoing [27]. A recent case study additionally identified a novel fusion of GAB1-ABL2 not previously reported in iHGG or any other cancer in a patient with a tumor methylation profile consistent with iHGG [33]. Based on this molecular analysis, dasatinib, an ABL inhibitor commonly used to treat CML, was trialed and found to be highly effective. This study highlights the impact of molecular profiling on treatment selection and outcomes. More recent studies have also shown that for these tumors, the traditional regimen, including radiation therapy, may not be needed depending on the effectiveness of molecular-targeted treatment options [31]. This is especially beneficial for overall survival and quality of life in the infant population, as molecular therapies have been shown to have fewer short- and long-term side effects.

2.5. NF1-Associated HGG and HGAP

Neurofibromatosis 1 (NF1) is an autosomal dominant disorder caused by a mutation in the NF1 tumor suppressor gene, classically associated with characteristic skin patches (café-au-lait macules) and neurofibromas, as well as Lisch nodules and freckling [34]. Pediatric patients with NF1 are most likely to develop low-grade gliomas, especially optic nerve gliomas, whereas NF1-associated tumors arising in adulthood are more likely high-grade gliomas [35]. Unsurprisingly, these tumors almost all harbor germline NF1 mutations, but further molecular classification of these tumors have identified other mutations that group these tumors into a distinct entity from other low-grade or high-grade gliomas [35]. Importantly, NF1-related tumors arising in both childhood and adulthood largely lack IDH mutations, unlike most pLGGs, and additionally do not contain histone 3 mutations, unlike many pHGGs [35,36]. A certain subset of NF1-associated tumors is also distinct in that they contain numerous CD3 and CD8-positive T-lymphocytes, indicating that this particular subtype of NF1-related tumors may respond favorably to immunotherapies [36].
High-grade gliomas occurring in NF1 typically fall between H3/IDH-WT tumors and a newly defined entity, high-grade astrocytoma with piloid features (HGAP), a subset of gliomas with an intermediate prognosis between low-grade astrocytomas and H3/IDH-WT pHGGs [37]. These tumors most commonly arise in adulthood and are most often located in the posterior fossa. In addition to NF1 germline and somatic mutations, this group is associated with alterations in MAPK pathway genes; as well as loss of function of ATRX, CDKN2A, and TP53; likening them to both pLGGs and pHGGs, respectively [37]. What truly distinguishes HGAP from other pLGGs or pHGGs is its methylation profile. NF1-associated HGAP is associated with dysregulation of RNA processing secondary to both hyper- and hypomethylation [37]. Improved understanding of the molecular and epigenetic profiles of these tumors has opened new avenues for treatment of gliomas in NF1. Inhibition of MEK, a component of the MAPK pathway, has been shown to be effective in some patients, as well as other MAPK and mTOR pathway inhibitors [38].

3. Discussion

As advances in molecular profiling of tumors become more widely available, differentiating pHGG into its molecular subtypes becomes increasingly crucial for both prediction of outcomes and treatment selection. The reliance on molecular identification is evident in the release of the WHO CNS5 guidelines in 2021, whose primary focus was on the addition of molecular profiling as part of the diagnostic criteria for these tumors. WHO CNS5 allowed for the identification of the H3G34-mutant, H3/IDH-WT, and iHGG as distinct from one another and H3K27-altered tumors for the first time in the WHO diagnostic criteria [39].
While traditional methods of histologic and immunohistochemical classification are beneficial in differentiating pHGG from pLGG, these differences are inadequate predictors of treatment response and long-term outcomes. Incorporation of next-generation sequencing (NGS) has become a crucial component of the diagnostic process at multiple institutions as a way to glean insight into the particular molecular and genetic alterations in individual tumors as a guide for both diagnosis and treatment [14]. NGS is both more time and cost efficient in addition to requiring smaller tissue samples sizes when compared to whole genome sequencing. However, this is not necessarily widely available at all institutions, and therefore more work is needed to identify the most cost effective and efficient way to identify molecular targets. Many institutions now use their own targeted CNS neoplasm NGS panels while other institutions submit tissue to third party vendors or outside hospitals. It will be essential that NGS and further updated technology is available for all patients to ensure the most up to date diagnostic information.
Current clinical trials seek to target these molecular differences such as histone mutations, specific methylation profiles, and gene fusions. This presents an opportunity to both tailor therapy to specific tumor subtypes and better understand prognosis. Further molecular classification of these tumors may provide insight as to appropriate methods of drug delivery for specific tumors, a significant barrier to efficacy of therapy in pHGG. Additionally, advances in targeted therapy for pHGG may allow for more limited use of chemotherapy and local radiation. While the current standard of treatment for pHGG remains resection and radiation therapy, upfront use of targeted therapy remains under investigation, particularly for its radiation sparing implications in iHGG [31,32]. Use of existing and novel targeted therapies, therefore, becomes an increasingly crucial component of pHGG treatment, either alone or in combination with traditional therapy. Further investigation into which patients and tumor subgroups may benefit most from targeted therapies is needed to develop a more specific standard of care for these tumors. Current studies have shown an overall improvement in outcomes with the addition of individualized therapies based on molecular and genetic findings. For example, a patient with iHGG who was treated with NTRK-inhibitors as first-line treatment in comparison to radiation has been shown to have stable disease control 3 years after therapy, resulting in the ability to avoid the adverse effects radiation and generalized chemotherapy altogether [40]. In H3G34-mutant tumors, a combination of PDGFRA inhibitors and mTOR inhibitors was shown to improve OS in these patients in comparison to traditional therapy [41]. An example of some ongoing clinical trials based on molecular alterations can be seen below (Table 2). This is continuously being updated with new data emphasizing the importance of ongoing pre-clinical work and its translation to clinical trials.

4. Conclusions

As the importance of individualized molecular, genetic, and epigenetic profiling in pediatric high-grade gliomas grows, it is crucial to understand the limitations of this new approach to diagnosis. Despite the improved ability to identify molecular targets in these tumors, the challenge of developing accurate pre-clinical models, as well as therapies that can effectively target the identified pathways and adequately cross the blood–brain barrier remains an issue [43,44]. There are also known disparities in access, making the inclusion of molecular profiling of tumors part of the new standard of care more difficult to achieve on a global scale. Studies have shown that patients of lower socioeconomic status are less likely to be offered or undergo methylation profiling, despite its emerging importance in both diagnostic and treatment decisions [45].
Regardless of these barriers, it is clear that identification of unique molecular and genetic targets in pHGG will allow for improvement in therapies and continued hope for a cure [46]. The emerging field of radiogenomics—using machine learning to predict molecular changes from a large database of common imaging findings—provides another avenue for future growth in the field [47].
While the prognosis for pHGG remains poor, a continued focus on directing pharmaceutical development towards specific molecular markers offers hope for improvement in OS and quality of life for patients with pHGG.

Author Contributions

Concept and design: E.V. and S.R. Drafting of the manuscript and critical revision: A.C., A.K., A.S., E.V. and S.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ABL2ABL proto-oncogene
ACVR1activin A receptor type 1
aHGGadult high-grade glioma
ALKanaplastic lymphoma kinase’
ATRXalpha-thalassemia/mental retardation, X-linked
AURKAAurora kinase A
BRAFV-raf murine sarcoma viral oncogene homolog B
CAR-Tchimeric antigen receptor T-cell therapy
CMLchronic myeloid leukemia
CNScentral nervous system
DHGdiffuse hemispheric glioma
DIPGdiffuse intrinsic pontine glioma
DMGdiffuse midline glioma
EGFRepidermal growth factor receptor
EZHIPenhancer of zeste homologs inhibitory protein
GAB1GRB2-associated binding protein 1
GFAPglial fibrillary acidic protein
H3K36me3trimethylated state of lysine 36 on histone 3
H3K27histone 3 mutation at lysine 27
H3G34histone 3 mutation at glycine 34
H3G34R/Vhistone 3 mutation at glycine 34, replacement with arginine/valine
H3/IDH-WThistone 3, isocitrate dehydrogenase wild-type
HDAChistone deacetylase
HGAPhigh-grade astrocytoma with piloid features
ID2inhibitor of DNA binding protein 2
iHGGinfant-type high-grade glioma
METMET proto-oncogene
MDM2murine double minute 2
MGMTO6-methylguanine-DNA methyltransferase
MRImagnetic resonance imaging
mTORmammalian target of rapamycin
MYCNmyelocytomatosis-neuroblastoma gene
NGSnext generation sequencing
NF1neurofibromatosis type 1
NTRKneurotrophic tyrosine receptor kinase
OLIG2oligodendrocyte transcription factor 2
OSoverall survival
PDGFRplatelet-derived growth factor receptor
pHGGpediatric high-grade glioma
PIK3R1phosphoinositide-3-kinase regulatory subunit 1
pLGGpediatric low-grade glioma
ROS1ROS proto-oncogene 1
RTKreceptor tyrosine kinase
SETD2SET domain containing 2
TERT promotertelomerase reverse transcriptase promoter
TP53tumor suppressor protein 53
WHOWorld Health Organization

References

  1. Graham, M.S.; Mellinghoff, I.K. Histone-Mutant Glioma: Molecular Mechanisms, Preclinical Models, and Implications for Therapy. Int. J. Mol. Sci. 2020, 21, 7193. [Google Scholar] [CrossRef]
  2. Diaz, A.K.; Baker, S.J. The Genetic Signatures of Pediatric High-Grade Glioma: No Longer a One-Act Play. Semin. Radiat. Oncol. 2014, 24, 240–247. [Google Scholar] [CrossRef]
  3. Park, Y.W.; Vollmuth, P.; Foltyn-Dumitru, M.; Sahm, F.; Ahn, S.S.; Chang, J.H.; Kim, S.H. 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 2023, 58, 690–708. [Google Scholar] [CrossRef]
  4. Guerra-García, P.; Marshall, L.V.; Cockle, J.V.; Ramachandran, P.V.; Saran, F.H.; Jones, C.; Carceller, F. Challenging the Indiscriminate Use of Temozolomide in Pediatric High-Grade Gliomas: A Review of Past, Current, and Emerging Therapies. Pediatr. Blood Cancer 2020, 67, e28011. [Google Scholar] [CrossRef] [PubMed]
  5. Perwein, T.; Giese, B.; Nussbaumer, G.; von Bueren, A.O.; van Buiren, M.; Benesch, M.; Kramm, C.M. How I Treat Recurrent Pediatric High-Grade Glioma (pHGG): A Europe-Wide Survey Study. J. Neurooncol. 2023, 161, 525–538. [Google Scholar] [CrossRef]
  6. Blasco-Santana, L.; Colmenero, I. Molecular and Pathological Features of Paediatric High-Grade Gliomas. Int. J. Mol. Sci. 2024, 25, 8498. [Google Scholar] [CrossRef]
  7. Gianno, F.; Giovannoni, I.; Cafferata, B.; Diomedi-Camassei, F.; Minasi, S.; Barresi, S.; Buttarelli, F.R.; Alesi, V.; Cardoni, A.; Antonelli, M.; et al. Paediatric-Type Diffuse High-Grade Gliomas in the 5th CNS WHO Classification. Pathologica 2022, 114, 422–435. [Google Scholar] [CrossRef] [PubMed]
  8. Chiang, J.; Diaz, A.K.; Makepeace, L.; Li, X.; Han, Y.; Li, Y.; Klimo, P.; Boop, F.A.; Baker, S.J.; Gajjar, A.; et al. Clinical, Imaging, and Molecular Analysis of Pediatric Pontine Tumors Lacking Characteristic Imaging Features of DIPG. Acta Neuropathol. Commun. 2020, 8, 57. [Google Scholar] [CrossRef] [PubMed]
  9. Das, S.; Ahlawat, S.; Panda, A.K.; Sarangi, J.; Jain, P.; Gupta, R.K.; Vaishya, S.; Patir, R. Pediatric High Grade Gliomas: A Comprehensive Histopathological, Immunohistochemical and Molecular Integrated Approach in Routine Practice. Pathol. Res. Pract. 2024, 258, 155347. [Google Scholar] [CrossRef]
  10. Tang, K.; Cesaire, M.; McDonald, T.; Cimino, P.J.; Castro, M.G.; Jackson, S. A Scoping Review of Diffuse Hemispheric Glioma, H3 G34-Mutant: Epigenetic and Molecular Profiles, Clinicopathology, and Treatment Avenues. Neurooncol. Adv. 2024, 6, vdae208. [Google Scholar] [CrossRef]
  11. Weinberg, D.N.; Allis, C.D.; Lu, C. Oncogenic Mechanisms of Histone H3 Mutations. Cold Spring Harb. Perspect. Med. 2017, 7, a026443. [Google Scholar] [CrossRef] [PubMed]
  12. Rao, S.; Sahay, A.; Epari, S. Paediatric Type Diffuse High Grade Gliomas in the WHO CNS5 Classification: What the Pathologist Needs to Know? Indian J. Pathol. Microbiol. 2022, 65, S50. [Google Scholar] [CrossRef]
  13. MacDonald, T.J.; Aguilera, D.; Kramm, C.M. Treatment of High-Grade Glioma in Children and Adolescents. Neuro-Oncol. 2011, 13, 1049–1058. [Google Scholar] [CrossRef]
  14. Miklja, Z.; Pasternak, A.; Stallard, S.; Nicolaides, T.; Kline-Nunnally, C.; Cole, B.; Beroukhim, R.; Bandopadhayay, P.; Chi, S.; Ramkissoon, S.H.; et al. Molecular Profiling and Targeted Therapy in Pediatric Gliomas: Review and Consensus Recommendations. Neuro-Oncol. 2019, 21, 968–980. [Google Scholar] [CrossRef]
  15. Grasso, C.S.; Tang, Y.; Truffaux, N.; Berlow, N.E.; Liu, L.; Debily, M.-A.; Quist, M.J.; Davis, L.E.; Huang, E.C.; Woo, P.J.; et al. Functionally-Defined Therapeutic Targets in Diffuse Intrinsic Pontine Glioma. Nat. Med. 2015, 21, 555–559. [Google Scholar] [CrossRef]
  16. Marini, B.L.; Benitez, L.L.; Zureick, A.H.; Salloum, R.; Gauthier, A.C.; Brown, J.; Wu, Y.-M.; Robinson, D.R.; Kumar, C.; Lonigro, R.; et al. Blood-Brain Barrier-Adapted Precision Medicine Therapy for Pediatric Brain Tumors. Transl. Res. 2017, 188, 27.e1–27.e14. [Google Scholar] [CrossRef]
  17. Monje, M.; Mahdi, J.; Majzner, R.; Yeom, K.W.; Schultz, L.M.; Richards, R.M.; Barsan, V.; Song, K.-W.; Kamens, J.; Baggott, C.; et al. Intravenous and Intracranial GD2-CAR T Cells for H3K27M+ Diffuse Midline Gliomas. Nature 2025, 637, 708–715. [Google Scholar] [CrossRef] [PubMed]
  18. Monje, M.; Vitanza, N.A.; Ronsley, R.; Choe, M.; Seidel, K.; Huang, W.; Rawlings-Rhea, S.D.; Beam, M.; Steinmetzer, L.; Wilson, A.L.; et al. Intracerebroventricular B7-H3-Targeting CAR T Cells for Diffuse Intrinsic Pontine Glioma: A Phase 1 Trial. Nat. Med. 2025, 31, 861–868. Available online: https://www.nature.com/articles/s41591-024-03451-3 (accessed on 17 April 2025).
  19. Korshunov, A.; Capper, D.; Reuss, D.; Schrimpf, D.; Ryzhova, M.; Hovestadt, V.; Sturm, D.; Meyer, J.; Jones, C.; Zheludkova, O.; et al. Histologically Distinct Neuroepithelial Tumors with Histone 3 G34 Mutation Are Molecularly Similar and Comprise a Single Nosologic Entity. Acta Neuropathol. 2016, 131, 137–146. [Google Scholar] [CrossRef]
  20. Nguyen, A.V.; Soto, J.M.; Gonzalez, S.-M.; Murillo, J.; Trumble, E.R.; Shan, F.Y.; Huang, J.H. H3G34-Mutant Gliomas—A Review of Molecular Pathogenesis and Therapeutic Options. Biomedicines 2023, 11, 2002. [Google Scholar] [CrossRef] [PubMed]
  21. Haase, S.; Nuñez, F.M.; Gauss, J.C.; Thompson, S.; Brumley, E.; Lowenstein, P.; Castro, M.G. Hemispherical Pediatric High-Grade Glioma: Molecular Basis and Therapeutic Opportunities. Int. J. Mol. Sci. 2020, 21, 9654. [Google Scholar] [CrossRef]
  22. Bonada, M.; Pittarello, M.; De Fazio, E.; Gans, A.; Alimonti, P.; Slika, H.; Legnani, F.; Di Meco, F.; Tyler, B. Pediatric Hemispheric High-Grade Gliomas and H3.3-G34 Mutation: A Review of the Literature on Biological Features and New Therapeutic Strategies. Genes 2024, 15, 1038. [Google Scholar] [CrossRef]
  23. Hong, L.; Shi, Z.-F.; Li, K.K.-W.; Wang, W.-W.; Yang, R.R.; Kwan, J.S.-H.; Chen, H.; Li, F.-C.; Liu, X.-Z.; Chan, D.T.-M.; et al. Molecular Landscape of Pediatric Type IDH Wildtype, H3 Wildtype Hemispheric Glioblastomas. Lab. Investig. 2022, 102, 731–740. [Google Scholar] [CrossRef]
  24. Korshunov, A.; Schrimpf, D.; Ryzhova, M.; Sturm, D.; Chavez, L.; Hovestadt, V.; Sharma, T.; Habel, A.; Burford, A.; Jones, C.; et al. H3-/IDH-Wild Type Pediatric Glioblastoma Is Comprised of Molecularly and Prognostically Distinct Subtypes with Associated Oncogenic Drivers. Acta Neuropathol. 2017, 134, 507–516. [Google Scholar] [CrossRef] [PubMed]
  25. Buccoliero, A.M.; Giunti, L.; Moscardi, S.; Castiglione, F.; Provenzano, A.; Sardi, I.; Scagnet, M.; Genitori, L.; Caporalini, C. Pediatric High Grade Glioma Classification Criteria and Molecular Features of a Case Series. Genes 2022, 13, 624. [Google Scholar] [CrossRef]
  26. Bender, K.; Kahn, J.; Perez, E.; Ehret, F.; Roohani, S.; Capper, D.; Schmid, S.; Kaul, D. Diffuse Paediatric-Type High-Grade Glioma, H3-Wildtype and IDH-Wildtype: Case Series of a New Entity. Brain Tumor Pathol. 2023, 40, 204–214. [Google Scholar] [CrossRef] [PubMed]
  27. Ceglie, G.; Vinci, M.; Carai, A.; Rossi, S.; Colafati, G.S.; Cacchione, A.; Tornesello, A.; Miele, E.; Locatelli, F.; Mastronuzzi, A. Infantile/Congenital High-Grade Gliomas: Molecular Features and Therapeutic Perspectives. Diagnostics 2020, 10, 648. [Google Scholar] [CrossRef] [PubMed]
  28. Stucklin, A.S.G.; Ryall, S.; Fukuoka, K.; Zapotocky, M.; Lassaletta, A.; Li, C.; Bridge, T.; Kim, B.; Arnoldo, A.; Kowalski, P.E.; et al. Alterations in ALK/ROS1/NTRK/MET Drive a Group of Infantile Hemispheric Gliomas. Nat. Commun. 2019, 10, 4343. Available online: https://pmc.ncbi.nlm.nih.gov/articles/PMC6761184/ (accessed on 8 May 2025).
  29. Clarke, M.; Mackay, A.; Ismer, B.; Pickles, J.C.; Tatevossian, R.G.; Newman, S.; Bale, T.A.; Stoler, I.; Izquierdo, E.; Temelso, S.; et al. Infant High-Grade Gliomas Comprise Multiple Subgroups Characterized by Novel Targetable Gene Fusions and Favorable Outcomes. Cancer Discov. 2020, 10, 942–963. [Google Scholar] [CrossRef]
  30. Chiang, J.; Bagchi, A.; Li, X.; Dhanda, S.K.; Huang, J.; Pinto, S.N.; Sioson, E.; Dalton, J.; Tatevossian, R.G.; Jia, S.; et al. High-Grade Glioma in Infants and Young Children Is Histologically, Molecularly, and Clinically Diverse: Results from the SJYC07 Trial and Institutional Experience. Neuro-Oncol. 2023, 26, 178–190. [Google Scholar] [CrossRef]
  31. Di Ruscio, V.; Carai, A.; Del Baldo, G.; Vinci, M.; Cacchione, A.; Miele, E.; Rossi, S.; Antonelli, M.; Barresi, S.; Caulo, M.; et al. Molecular Landscape in Infant High-Grade Gliomas: A Single Center Experience. Diagnostics 2022, 12, 372. [Google Scholar] [CrossRef]
  32. Lassaletta, A.; Guerreiro Stucklin, A.; Ramaswamy, V.; Zapotocky, M.; McKeown, T.; Hawkins, C.; Bouffet, E.; Tabori, U. Profound Clinical and Radiological Response to BRAF Inhibition in a 2-Month-Old Diencephalic Child with Hypothalamic/Chiasmatic Glioma. Pediatr. Blood Cancer 2016, 63, 2038–2041. [Google Scholar] [CrossRef] [PubMed]
  33. Buckner-Wolfson, E.; Jung, G.; Kim, T.; Fatemi, R.; Liriano, G.; Dalvi, N.; Behbahani, M.; Chin, S.; Martin, A.; Kobets, A. A Case Report of Infant-Type Hemispheric Glioma with a Novel GAB1-ABL2 Kinase Fusion Treated with Dasatinib. Pediatr. Neurosurg. 2024, 59, 27–34. [Google Scholar] [CrossRef]
  34. Peduto, C.; Zanobio, M.; Nigro, V.; Perrotta, S.; Piluso, G.; Santoro, C. Neurofibromatosis Type 1: Pediatric Aspects and Review of Genotype–Phenotype Correlations. Cancers 2023, 15, 1217. [Google Scholar] [CrossRef]
  35. D’Angelo, F.; Ceccarelli, M.; Tala; Garofano, L.; Zhang, J.; Frattini, V.; Caruso, F.P.; Lewis, G.; Alfaro, K.D.; Bauchet, L.; et al. The Molecular Landscape of Glioma in Patients with Neurofibromatosis 1. Nat. Med. 2019, 25, 176–187. [Google Scholar] [CrossRef]
  36. Packer, R.J.; Iavarone, A.; Jones, D.T.W.; Blakeley, J.O.; Bouffet, E.; Fisher, M.J.; Hwang, E.; Hawkins, C.; Kilburn, L.; MacDonald, T.; et al. Implications of New Understandings of Gliomas in Children and Adults with NF1: Report of a Consensus Conference. Neuro-Oncol. 2020, 22, 773–784. [Google Scholar] [CrossRef] [PubMed]
  37. Cimino, P.J.; Ketchum, C.; Turakulov, R.; Singh, O.; Abdullaev, Z.; Giannini, C.; Pytel, P.; Lopez, G.Y.; Colman, H.; Nasrallah, M.P.; et al. Expanded Analysis of High-Grade Astrocytoma with Piloid Features Identifies an Epigenetically and Clinically Distinct Subtype Associated with Neurofibromatosis Type 1. Acta Neuropathol. 2023, 145, 71–82. [Google Scholar] [CrossRef]
  38. Lucas, C.-H.G.; Sloan, E.A.; Gupta, R.; Wu, J.; Pratt, D.; Vasudevan, H.N.; Ravindranathan, A.; Barreto, J.; Williams, E.A.; Shai, A.; et al. Multiplatform Molecular Analyses Refine Classification of Gliomas Arising in Patients with Neurofibromatosis Type 1. Acta Neuropathol. 2022, 144, 747–765. [Google Scholar] [CrossRef]
  39. Louis, D.N.; Perry, A.; Wesseling, P.; Brat, D.J.; Cree, I.A.; Figarella-Branger, D.; Hawkins, C.; Ng, H.K.; Pfister, S.M.; Reifenberger, G.; et al. The 2021 WHO Classification of Tumors of the Central Nervous System: A Summary. Neuro-Oncol. 2021, 23, 1231–1251. [Google Scholar] [CrossRef]
  40. Simoneau, J.; Robertson, P.; Muraszko, K.; Maher, C.O.; Garton, H.; Calvert, R.; Koschmann, C.; Upadhyaya, S.A.; Mody, R.; Brown, N.; et al. Long-Term Tumor Stability After First-Line Treatment with Larotrectinib in an Infant with NTRK2 Fusion–Positive High-Grade Glioma. JNCCN 2024, 22. [Google Scholar] [CrossRef]
  41. Miklja, Z.; Yadav, V.N.; Cartaxo, R.T.; Siada, R.; Thomas, C.C.; Cummings, J.R.; Mullan, B.; Stallard, S.; Paul, A.; Bruzek, A.K.; et al. Everolimus Improves the Efficacy of Dasatinib in PDGFRα-Driven Glioma. J. Clin. Investig. 2020, 130, 5313–5325. [Google Scholar] [CrossRef] [PubMed]
  42. Xi, X.; Liu, N.; Wang, Q.; Chu, Y.; Yin, Z.; Ding, Y.; Lu, Y. ACT001, a Novel PAI-1 Inhibitor, Exerts Synergistic Effects in Combination with Cisplatin by Inhibiting PI3K/AKT Pathway in Glioma. Cell Death Dis. 2019, 10, 757. [Google Scholar] [CrossRef] [PubMed]
  43. Porter, A.B.; Wen, P.Y.; Polley, M.-Y.C. Molecular Profiling in Neuro-Oncology: Where We Are, Where We’re Heading, and How We Ensure Everyone Can Come Along. Am. Soc. Clin. Oncol. Educ. Book 2023, 43, e389322. [Google Scholar] [CrossRef]
  44. McFaline-Figueroa, J.R.; Wen, P.Y. Negative Trials over and over Again: How Can We Do Better? Neuro-Oncol. 2022, 25, 1–3. [Google Scholar] [CrossRef]
  45. Malmström, A.; Łysiak, M.; Kristensen, B.W.; Hovey, E.; Henriksson, R.; Söderkvist, P. Do We Really Know Who Has an MGMT Methylated Glioma? Results of an International Survey Regarding Use of MGMT Analyses for Glioma. Neuro-Oncol. Pract. 2020, 7, 68–76. [Google Scholar] [CrossRef]
  46. Grigore, F.-N.; Yang, S.J.; Chen, C.C.; Koga, T. Pioneering Models of Pediatric Brain Tumors. Neoplasia 2023, 36, 100859. [Google Scholar] [CrossRef] [PubMed]
  47. Madhogarhia, R.; Haldar, D.; Bagheri, S.; Familiar, A.; Anderson, H.; Arif, S.; Vossough, A.; Storm, P.; Resnick, A.; Davatzikos, C.; et al. Radiomics and Radiogenomics in Pediatric Neuro-Oncology: A Review. Neuro-Oncol. Adv. 2022, 4, vdac083. [Google Scholar] [CrossRef] [PubMed]
Table 1. Summary of the four pHGG subtypes as defined by the WHO CNS5 2021 guidelines and their associated molecular and genetic changes.
Table 1. Summary of the four pHGG subtypes as defined by the WHO CNS5 2021 guidelines and their associated molecular and genetic changes.
Tumor TypeKey Molecular and Genetic Changes
H3K27-altered gliomaTP53, ACVR1, PDGFRA, EGFR, EZHIP
H3G34-altered gliomaTP53, ATRX, PDGFRA, MYCN
H3/IDH-WT gliomaTP53, PDGFRA, EGFR, MYCN, ID2, IDH-WT, H3-WT
iHGGNTRK, ALK, ROS, MET
NF1/HGAPMAPK, ATRX, CDK2A, TP53
Table 2. Ongoing clinical trials regarding pHGG.
Table 2. Ongoing clinical trials regarding pHGG.
Clinicaltrials.gov IDBrief TitlePhase/StatuspHGG TypeDrug Type Target
NCT06838676 ACT001 for the Treatment of Diffuse Intrinsic Pontine Gliomas and H3K27-altered High Grade GliomasPhase IIH3K27-altered gliomaPAI-1 inhibitor, inhibits glioma cell proliferation [42]
NCT06333899 Lorlatinib for Newly Diagnosed High-Grade Glioma With ROS or ALK Fusion Early Phase IH3K27-altered glioma, iHGGALK inhibtor
NCT06946680 IL-8 Receptor-modified CD70 CAR T Cell Therapy in CD70+ Pediatric High-grade Glioma (HGG) Phase INew diagnosis pHGGCD70+ HGG immunotherapy
NCT06712875 MAPK Inhibition Combined with Anti-PD1 Therapy for BRAF-altered Pediatric GliomasPhase I/Phase IIpLGG, pHGGMEK1/2 inhibition, PD-1 inhibition, BRAF kinase inhibitor
NCT05610891 Novel Targeted Radiotherapy in Pediatric Patients With Inoperable Relapsed or Refractory HGG Phase IRelapsed, refractory pHGG
NCT04911621 Adjuvant Dendritic Cell Immunotherapy for Pediatric Patients With High-grade Glioma or Diffuse Intrinsic Pontine GliomaPhase 1/Phase 2pHGG
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Vallee, E.; Steller, A.; Childress, A.; Koch, A.; Raskin, S. The Current Landscape of Molecular Pathology for the Diagnosis and Treatment of Pediatric High-Grade Glioma. J. Mol. Pathol. 2025, 6, 17. https://doi.org/10.3390/jmp6030017

AMA Style

Vallee E, Steller A, Childress A, Koch A, Raskin S. The Current Landscape of Molecular Pathology for the Diagnosis and Treatment of Pediatric High-Grade Glioma. Journal of Molecular Pathology. 2025; 6(3):17. https://doi.org/10.3390/jmp6030017

Chicago/Turabian Style

Vallee, Emma, Alyssa Steller, Ashley Childress, Alayna Koch, and Scott Raskin. 2025. "The Current Landscape of Molecular Pathology for the Diagnosis and Treatment of Pediatric High-Grade Glioma" Journal of Molecular Pathology 6, no. 3: 17. https://doi.org/10.3390/jmp6030017

APA Style

Vallee, E., Steller, A., Childress, A., Koch, A., & Raskin, S. (2025). The Current Landscape of Molecular Pathology for the Diagnosis and Treatment of Pediatric High-Grade Glioma. Journal of Molecular Pathology, 6(3), 17. https://doi.org/10.3390/jmp6030017

Article Metrics

Back to TopTop