All abstracts are listed here (both oral and poster presentations) by the alphabetical order of the surnames of the first author.

 

Aldape, Kenneth

National Cancer Institute

The NCI/Bethesda DNA Methylation Classifier: an Important Diagnostic Tool for CNS Tumors

 

DNA methylation-based classification has become essential for accurately diagnosing CNS tumor subtypes, by providing a reproducible and objective tool in addition to existing diagnostic modalities. While the commonly used Heidelberg classifier is highly accurate and valuable, we find utility in creating an alternative classifier to 1) provide a ‘second look’ at the methylation results in addition to the Heidelberg classifier; and 2) offer the flexibility to add recently described classes to the classifier. To this end, the NCI/Bethesda classifier version 3.0 was developed, starting with a set of 19,642 samples, representing 201 unique tumor types/subtypes. We then split this reference set into an 80% training set (n=15,467) and a 20% internal validation set (n=3,800). The 15,467-sample training set served as the foundation for building hierarchical Support Vector Machine (SVM) classifiers. In the first stage, we classified samples into 22 major tumor superfamilies. In the second layer of classification, we further subclassified each superfamily into specific tumor types/subtypes. We compared the results of this classifier with existing sample labels that had been derived from a previous version of the NCI/Bethesda classifier, as well as from the Heidelberg version 12.8 classifier. At the individual class level, mean accuracies ranged between 97% and 99%. Our first level of model performance evaluation was based on cross-validation accuracies, and we extended this to predict superfamily and class for the 20% internal validation set(n=3800), showing 95.6% accuracy (3,633 correctly classified out of 3,800 samples), using a 0.9 confidence score threshold. We then applied both the Heidelberg classifier and the NCI/Bethesda v3.0 classifier to an external validation set n=1963 samples), which was completely unseen at the time of model creation. With the 0.90 threshold cutoff, the DKFZ classifier correctly classified only 1,033 out of 1,963 samples, achieving a match rate of 52.6%. In contrast, the NCI/Bethesda classifier correctly classifying 1,383 out of the 1,963 samples, achieving a higher match rate of 70.5%. High-confidence matches were reached in both classifiers for 836/1,963 samples. A high-confidence match to only the Heidlberg classifier was reached in 197 samples, while 547 samples showed a high-confidence match to only the NCI/Bethesda classifier, highlighting the potential utility of using both classifiers in daily practice. Two tumor types in the current WHO CNS classification, absent from the Heidelberg classifier (Desmoplastic myxoid tumor of the pineal region, SMARCB1-mutant, and Intracranial mesenchymal tumor, FET::CREB fusion-positive) are present in the NCI/Bethesda classifier, enabling their identification in suspected cases. In addition, newly described classes, including Dural angioleiomyoma and High‑grade glioma with pleomorphic and pseudopapillary features, as well as others, are included in the NCI/Bethesda classifier. Use of our classifier is freely available following simple registration at https://methylscape.ccr.cancer.gov/. In sum, the NCI/Bethesda provides a tool that can be used to robustly classify CNS tumors, providing an alternative machine learning method as well as additional tumor types relative to the Heidelberg classifier. Our current consultation practice employs both classifiers, in an effort to fully utilize the merits of each.

 

 

Andriani, Rini

Dharmais National Cancer Center of Indonesia

Association of Isocitrate Dehydrogenase (IDH) Mutation Status with Seizure Incidence in Glioma Patients: Insights from the Indonesian Cancer Center in 2023-2024

 

Background:

Seizures are a common clinical manifestation in glioma patients and, if uncontrolled, can significantly contribute to morbidity, cognitive decline, and a diminished quality of life. Previous studies have suggested an association between lower-grade gliomas and a higher incidence of seizures, but the underlying mechanisms remain unclear.

Objective: This study aims to identify the roles of isocitrate dehydrogenase (IDH) biomarker mutations for clinical seizure prediction in glioma patients in Indonesia.

 

Methods:

The study design is retrospective based on the data collected at Dharmais National Cancer Center Hospital, Jakarta, Indonesia. We examined all glioma patients admitted to our hospital between January 1, 2023, and October 1, 2024. All glioma patients were confirmed by histopathological analysis. We examined the association between IDH mutation status and seizure incidence during hospitalization or admission. We collected data on the demographic and clinical characteristics of patients on age, sex, glioma type (astrocytoma or oligodendroglioma), WHO grade (2-4), tumor location (supratentorial or infratentorial), and treatment history. Statistical analyses were performed using Stata version 17.0 to estimate the odds ratio (OR) to represent the degree of association between seizure and IDH using logistic regression. Multivariable logistic regression was used to control for potential confounders.

 

Result:

Seizures were observed in 42 of the 66 patients (63.6%); among these, 27 patients (64.3%) had the IDH R132H mutation with a statistically significant association (p < 0.05). Glioma patients with mutated IDH R132H had 3.58 higher odds of experiencing seizures than those with the IDH wild-type (OR = 3.58, 95% CI: 1.07—12.0, p < 0.05), after adjusting to sex and age variable. All patients with oligodendroglioma experienced seizures (p = 0.05). Patients over 50 years old had a lower risk of seizure occurrence compared to patients under 40 (OR = 0.22, 95% CI: 0.063 — 0.76, p < 0.05), while no statistically significant difference was observed for the 40-50 age group (OR = 2.70, 95% CI: 0.39-- 18.8, p > 0.05). Sex was not a statistically significant predictor of seizure risk in glioma patients (male vs. female OR = 2.20, 95% CI: 0.66 —7.36, p > 0.05). Analysis of tumor grade showed no statistically significant difference in seizure incidence among WHO tumor grades 2, 3, and 4. Grade 3 tumors had a higher but non-significant OR for seizures compared to Grade 2 (OR = 3.85, 95% CI: 0.93—15.86, p > 0.05).

 

Conclusion:

IDH mutation status is a significant predictor of seizure incidence in glioma patients, compared to wild-type IDH. The age over 50 has a lower risk of seizures than the age below 40, while the age group 40-50 does not demonstrate significant predictive power. These findings highlight the importance of the clinical prediction roles of IDH mutations and younger age in the treatment of glioma-related epilepsy.

 

Keywords: Indonesia, glioma, seizures, isocitrate dehydrogenase (IDH), epilepsy

 

 

Chang, Qing

Beijing Neurosurgical Institute, Bejing Tiantan Hospital

Tumor associated macrophage derived exosomes modulate immunotherapeutic sensitivity of SHH medulloblastoma by m6A modification

 

Background:

Medulloblastoma (MB) is the most common malignant brain tumor in children. The infiltration of tumor-associated macrophages (TAMs) in tumor microenvironment (TME) is correlated with the poor prognosis in SHH subtype of MB patients. Epigenetic alterations on RNA m6A modification were identified in this tumor. However, it remains unclear whether the TAMs infiltration in TME was correlated with the m6A modification status during tumor progression.

 

Methods:

The expression of m6A modification-related proteins and TAMs specific marker were assessed by immunohistochemistry in 40 SHH MB primary tumors. Methylated RNA immunoprecipitation sequencing (MeRIP-seq) and transcriptomic RNA sequencing (RNA-seq) were used to screen target genes affected by TAM-derived exosomes in SHH MB cells. Mechanisms of m6A-modification on target gene (FOXD1) were demonstrated by WB, qPCR, RNA half-life time and RNA immunoprecipitation assays. Combination treatment with AAV2/9-shFOXD1 and PD-1 inhibitors were performed in the NeuroD2: SmoA1 mouse model.

 

Results:

We reported for the first time that significant infiltration of TAMs was associated with decreased expression of METTL14 in primary SHH MBs. Exosomal microRNAs (miR-320e, miR-196b-5p, and miR-628-5p) released by TAMs targeted to the 3’-UTR (3’ Untranslated Region) of METTL14 in MB cells, promoting the gene’s degradation and consequently downregulating the overall m6A level in SHH MB. MeRIP-seq and RNA-seq analyses screened out Forkhead box D1 (FOXD1) as the target gene upregulated by TAMs-derived exosomes through m6A modification in MB cells, which was correlated with poor prognosis in SHH MB patients. Further experiments in vitro verified YTHDF2 can recognize m6A-modified FOXD1 and promote its mRNA degradation. Knockdown of FOXD1 in SHH MB cells significantly promoted the release of the CD8+T cell recruiting chemokines CXCL10/11. Treatment with AAV2/9-shFOXD1 significantly enhanced the anti-tumor effect of PD-1 inhibitor in NeuroD2: SmoA1 SHH MB mice.

 

Conclusion:

Our study revealed the m6A modification status of SHH MBs influenced by TAMs derived exosomal miRNAs. FOXD1 promoted the malignant progression of SHH MB through m6A modification. Increased immunotherapeutic sensitivity of the combination treatment with FOXD1 knockdown and anti-PD-1 blockade in vivo shed lights on a novel therapeutic strategy of SHH MB.

 

 

Chen, Bo

The University of Hong Kong

SIRT5 Suppresses Tumor Growth by Regulating Mitochondrial Metabolism and Synaptic Remodeling in Gliomas

 

Sirtuin 5 (SIRT5) is increasingly recognized as a key regulator of cellular metabolism, which is commonly dysregulated in cancer cells, resulting in enhanced proliferation and tumor progression. To investigate the clinicopathologic implications of SIRT5 dysregulation in glioblastoma, we performed comprehensive analyses of transcriptomic data and functional verifications using in vitro and in vivo glioblastoma models. We found that higher SIRT5 expression levels were associated with a favorable prognosis in glioma patients. Knockdown of SIRT5 significantly enhanced glioblastoma cell growth. Our data suggest its potential role in regulating mitochondrial metabolism in gliomas. Furthermore, SIRT5 is also significantly correlated with synaptic remodeling pathways. Our findings indicate a tumor-suppressive role for SIRT5 that extends beyond regulating cancer metabolism by which it may function through modulating neuroplasticity. Understanding these cellular interactions provides nuanced insights into the multifaceted role of SIRT5 and the broader therapeutic implications for the development of novel treatment strategies.

 

 

Chen, Zhilin

The University of Hong Kong

Single-nucleus and spatial transcriptomics of intracranial germinoma

 

Intracranial germinoma is the most common pediatric CNS germ cell tumour. While the survival rates of patients following standard treatment with radiotherapy plus carbo-etoposide are high, the children inevitably suffer from long-term cognitive impairment and neurological damage due to craniospinal irradiation. Germinomas often exhibit substantial lymphocyte infiltration; however, as they lack expression of immune checkpoint proteins, making them unlikely candidates for PD-1 or CTLA-4 blockage therapy. Indeed, the roles of the immune infiltrates in the tumour microenvironment of germinoma are poorly understood.


Therefore, we aim to characterize the tumour microenvironment of intracranial germinomas using single-nucleus RNA sequencing (snRNA-seq) and spatial transcriptomics (ST). After quality control, barcode filtering, and doublet/multiplet removal, we annotated single nuclei by the expression of marker genes, and integrated the annotated nuclei across multiple patients while ensuring the removal of batch effect. As expected, we identified substantial populations of T cells, B cells, and myeloid cells. Moreover, the germinoma cells express OCT4/POU5F1, KIT, and PDPN, while lacking expression of CD30 and AFP, consistent with the histological diagnostic criteria for germinoma. We further characterized the copy-number and cytogenetic alterations in the germinoma cells. We identified potential ligand-receptor communications between immune and germinoma cells. Additionally, we mapped the single nuclei to the spatial transcriptomic data and identified immune niches in the tumour microenvironment.. Finally, we will replicate our findings on a larger cohort of germinoma.

We hope that understanding the immune-tumour interactions will lead to the development of less cytotoxic treatments for patients with germinoma. This could potentially de-escalate or supplant craniospinal irradiation, enhancing the quality of life for the patients, while preserving survival outcomes.

 

 

Chong, Kyuha

Samsung Medical Center

The Role of Extra-Domain B Fibronectin in Glioblastoma Cell Invasion

 

Glioblastoma (GBM) is a highly aggressive tumor marked by its resistance to current treatments and high recurrence rates. Extra-domain B Fibronectin (EDB-FN), a glycoprotein absent in normal tissues but selectively expressed in the tumor microenvironment, was previously identified as a promising molecular marker for brain tumors, with potential applications in prognostics and therapeutic targeting. Despite this, the specific role and mechanism of EDB-FN in GBM cells remain undefined. In this study, the role and mechanisms of EDB-FN in contributing to GBM malignancy were investigated through in vitro experiments. The results demonstrate that silencing EDB-FN in GBM cells significantly reduces cell migration, invasiveness, extracellular matrix adhesion, and stem cell-like properties. Moreover, it was identified that the FAK and ERK signaling pathways are crucial for GBM cell progression in response to EDB-FN. This study highlights the potential of EDB-FN as a specific biomarker for GBM treatment, paving the way for future research in this area.

 

 

Clautrier, Simon

GH Saintes-Saint Jean d'Angely

Potentiation of treatments for glioblastoma: Analysis of a repositioning strategy for Metformin, Simvastatin and Valproic Acid associated with the Stupp protocol

 

Glioblastoma (GBM) is the most aggressive primary glial brain tumor in adults. Despite the Stupp protocol, prognosis remains poor, with overall survival of 14.6 months. The mechanisms of resistance are multifactorial: the importance of epigenetic modulations, the persistence of stem cells and the adoption by the cell of an aerobic glycolytic metabolism (Warburg effect) are elements that have been considered to be major causes of radiochemical resistance and short/medium-term failure of standard treatment.

This study explores the potential repositioning of Valproic Acid (VPA), Metformin, and Simvastatin in GBM treatment, evaluating their ability to target resistance mechanisms and potentiate Temozolomide's (TMZ) effect.

 

Material and Method:

A review of the literature analysed the mechanisms of action of these molecules on the radiochemical resistance mechanisms described

 

Results:

An analysis of the literature reveals that :

On GBM epigenetic modulations:

VPA, as a histone deacetylase inhibitor (HDACi), modifies gene expression patterns. VPA decreases O6-methylguanine-DNA methyltransferase (MGMT) expression and induces methylation of the MGMT promoter, potentially resensitizing TMZ-resistant GBM cells

MicroRNA-21 (miR-21) is overexpressed in 44-100% of GBM, contributing to chemoresistance. miR-21 is remarkably down-regulated by Metformin in a dose- and time-dependent manner in certain cancer cell lines found in the literature

On GBM cellular metabolism:

w   Metformin and VPA target the Warburg effect by activating AMP-activated protein kinase (AMPK), reducing tumor growth and promoting apoptosis.

On GBM signaling pathways:

w   Liver Kinase B1 (LKB1)/AMPK/mammalian Target Of Rapamycin (mTOR) pathway: Metformin activates AMPK, inhibiting mTOR.

w   HIPPO/Yes-Associated Protein (YAP)/Transcriptional co-Activator with PDZ-binding motif (TAZ) pathway: Simvastatin reduces YAP protein activity by inhibiting mevalonate synthesis.

w   FOS Like 1, AP-1 Transcription Factor Subunit (FOSL1): VPA could indirectly inhibit FOSL1 by increasing miR-138 expression.

On cancer stem cells:

All three molecules show potential for targeting GBM stem cells (GSCs) by modulating stem cell markers and signaling pathways.

On tumor suppressor gene p53:

w   Metformin and VPA could impact p53 reactivation, with potentially superior efficacy on cells with functional p53. VPA/Metformin: synergistic p53-dependent action

w   Simvastatin: preferential action on mutated p53

w   The triple combination could be relevant regardless of tumour heterogeneity The favourable physicochemical properties (liposolubility), low cost and good general tolerance of these compounds are additional advantages.

Discussion/Conclusion:

The combination of VPA, Simvastatin, and Metformin offers a multi-targeted approach to GBM treatment by:

w   Reversing epigenetic alterations, including MGMT methylation, and modulating microRNA expression.

w   Disrupting cancer cell metabolism through the Warburg effect.

w   Interfering with key signaling pathways involved in tumor growth and survival.

w   Targeting GBM stem cells.

w   Potentially reactivating tumor suppressor functions, particularly p53.

This strategy could address the limitations of single-molecule approaches in the face of GBM's heterogeneity. The synergistic effects of these drugs, combined with their ability to potentiate TMZ treatment, present a promising avenue for improving GBM therapy.

The study suggests that this combination could improve the efficacy of standard treatment and potentially overcome resistance mechanisms.

However, further preclinical research is required to elucidate the interactions between these molecules and validate their combined effects on GBM cells before proceeding with clinical trials.

 

 

Cui, Xiaoteng

Tianjin Medical University General Hospital

The basic and clinical research of targeting EGFR and mevalonate pathways synergize the antitumor effect of temozolomide in glioblastoma

 

Background:

Glioblastoma (GBM) represents the most malignant primary tumor in the central nervous system. GBM patients endure rapid progression and poor prognosis, with a median survival time of 15 months, despite surgical resection combined with radio- and temozolomide-based chemo-therapy since the first diagnosis. Metabolic reprogramming is a remarkable hallmark of GBM, not only producing enough energy for highly proliferative tumor cells, but also contributing to immunosuppressive tumor microenvironment formation. Epidermal growth factor receptor (EGFR) amplification and EGFR-vIII constitutively activated mutation are often detected in GBM cells, playing vital roles in the malignant behavior. Emerging evidences have suggested that hyper-activated EGFR pathway promotes to lipogenesis and cholesterol uptake. However, whether the EGFR pathway contributes to fatty acid metabolism and energy generation remains mystery.

 

Material and Methods:

Single-cell RNA sequencing, bulk RNA sequencing, and untargeted metabolomics analysis of fresh GBM specimens were performed. Mitochondrial function was detected by seahorse XF analyzer in GBMs with inhibiting EGFR pathway transductions. Orthotopic GBM models were constructed in immunodeficient nude mice and immunocompetent C57BL/6J mice.

 

Results:

GBMs with high expression of EGFR had characteristics of lipid remodeling and maintaining high cholesterol levels. A metabolism-associated RTK-fatty acid-gene signature (RFA) was well-constructed to predict the prognosis of GBM patients. Inhibition of the EGFR pathway and mevalonate pathways could remodel energy metabolism by repressing the tricarboxylic acid cycle and modulating ATP production. Mechanistically, hyper-activated EGFR pathway promoted the expressions of ACSS3, ACSL3, and ELOVL2 in an NF-κB-dependent manner. Moreover, blockade of mevalonate pathway reduced the EGFR level on the cell membranes, thereby affecting the signal transduction of the EGFR/AKT pathway. Targeting EGFR pathway and mevalonate pathways enhanced the antitumor effect of temozolomide in vitro and in vivo. Furthermore, GBM patients with the treatment of atorvastatin and temozolomide showed a prolonged progression free survival (PFS) time and overall survival (OS) time.

 

Conclusion:

This study uncovered a regulatory mechanism by which EGFR pathway activation promotes energy metabolism by upregulating ACSS3, ACSL3, and ELOVL2 expressions in GBMs. The combinatorial therapeutic strategy of temozolomide, auxiliary EGFR-TKI, and statins can benefit GBM patients with high RFA scores by significantly prolonging their survival time. Now the phase II clinical trials of temozolomide and atorvastatin against GBM is in the recruiting status (NCT06327451).

 

 

Dong, Jun

The Second Affiliated Hospital of Soochow University

Repurposing of amlodipine against glioma stem cells through regulating CD133–EGFR/AKT/mTOR cascade to downregulate SOX2 expression

 

Background:

Glioblastoma is the most aggressive and lethal primary brain tumor in adults with poor prognosis. Previous studies attribute high therapeutic resistance and short-term recurrence due to existence of glioma stem cells (GSCs), a valuable target for specific effective therapy against glioblastoma. Amlodipine is a classical calcium channel blocker (CCB) with exact anti-tumor effect independent of CCB activitywhich has potential of repurposing against glioblastoma.

 

Methods:

Cell viability, proliferation, self-renewal, invasive, apoptosis of GSCs were evaluated with CCK-8, EdU incorporation, tumor sphere formation assay, 3D spheroid invasion assay after administration of amlodipine. Flow cytometry was applied to detect apoptosis of GSCs, and Western blot assay was performed to explore the relevant protein levels involved in molecular mechanisms of amlodipine against GSCs. The interrelationship between GSC markers and key proteins of EGFR/Akt/mTOR signaling pathway was investigated by bioinformatics assay with GEPIA and further verified by Western blot. Exogenous activation of AKT phosphorylation by adding AKT agonist SC79 to evaluate the role of Akt signaling in the regulation of glioma stemness. The effect of amlodipine against GSCs was verified in intracranial orthotopic GSCs-derived tumor model, bioluminescence imaging was applied to monitor tumors growth in vivo, Ki67 staining was performed to detect proliferation of GSCs in vivo, TUNEL staining was used to detect apoptosis and expression of GSC markers in situ was identified by western blot and immunohistochemistry.

 

Results:

The present study disclosed that amlodipine inhibited cell viability, suppressed self-renewal, down-regulated stemness, induced apoptosis of GSCs obviously, and significantly reduced GSCs-derived intracranial tumor growth, prolonged survival of tumor-bearing mice as well. Mechanistically, amlodipine decreased expression of GSC-specific marker CD133. CD133 deletion destabilized epidermal growth factor receptor (EGFR) by enhancing EGFR internalization, thereby suppressing EGFR/Akt/mTOR signaling, leading to down-regulation of core transcriptional regulator SOX2 expression, and exogenous activation of AKT phosphorylation by adding AKT agonist SC79 partially reversed the antitumor effects of amlodipine. Besides, other dihydropyridine CCBs nicardipine and nifedipine as well as Ca2+ chelator BAPTA-AM did not reveal inhibitory effect against GSCs, L-type Ca2 + channel agonist also did not attenuate the inhibitory effect of amlodipine on GSCs, implying existence of the exact effect of amlodipine against GSCs.

 

Conclusions:

Amlodipine suppressed GSCs-initiated tumor development via regulating CD133 expression to inhibit EGFR/Akt/mTOR cascade to downregulate SOX2, indicating amlodipine had novel potential targeting on GSCs against glioblastoma.

 

 

Dong, Yijun

Peking Union Medical College

Single cell analysis of multifocal GBM reveals the function of ferroptosis induced NUPR1+ BMDM in promoting natural evolution

 

Glioblastoma (GBM) is an aggressive brain tumor with high levels of heterogeneity, contributing to its resistance to treatment and poor prognosis. Multifocal GBM, characterized by the presence of two or more distinct tumor foci within the same patient, presents a unique model for studying parallel tumor evolution. In this study, we investigated the role of bone marrow-derived macrophages (BMDMs) in the natural evolution of multifocal GBM using single-cell RNA sequencing (scRNA-seq), single-cell ATAC sequencing (scATAC-seq) and spatial transcriptomics from multiple patient samples.

We collected data from four patients with multifocal GBM, analyzing both older and younger lesions. By comparing copy number variation (CNV) and performing pseudotime trajectory analysis, we established two distinct evolutionary trajectories, defining the older lesions as highly evolved and the younger lesions as less evolved. Tumor subtypes, including mesenchymal (MES)-like tumor cells, were found to increase in the highly evolved lesions, which were enriched with a specific subpopulation of BMDMs expressing NUPR1. These NUPR1+ BMDMs localized predominantly to necrotic regions of the tumor, areas known to harbor hypoxia and cell death, and were associated with poor patient prognosis.

 

Further analysis revealed that the NUPR1+ BMDM subpopulation was related to ferroptosis, a form of regulated cell death driven by iron-dependent lipid peroxidation. The NUPR1+ BMDMs exhibited elevated ferroptosis-related gene expression, indicating that they may play a role in promoting this form of cell death within the tumor microenvironment. Spatial transcriptomic analysis also showed that SPP1 signaling between MES tumor cells and NUPR1+ BMDMs was enriched in necrotic regions, suggesting a crosstalk mechanism that drives tumor progression.

 

To validate these findings, we conducted in vitro experiments using the THP-1 macrophage cell line. We knocked down NUPR1 expression in these cells using siRNA and observed a significant decrease in the proliferation and migration of co-cultured GBM cells. Additionally, the expression of MES markers in the tumor cells was reduced, indicating that NUPR1+ BMDMs may promote the mesenchymal-epithelial transition (MET) phenotype, which is linked to increased malignancy in GBM.

 

Overall, our results suggest that NUPR1+ BMDMs contribute to the natural evolution of GBM by facilitating ferroptosis and promoting mesenchymal transition, particularly in necrotic areas. This subpopulation of macrophages represents a potential target for therapeutic intervention, as disrupting their function could inhibit tumor progression and improve patient outcomes. Future studies will further explore the mechanisms of NUPR1 in ferroptosis and its interactions with other immune and tumor cell populations in the GBM microenvironment.

 

 

Duan, Zejun

Sanbo Brain Hospital, Capital Medical University, Beijing

Rare DICER1-mutant primary intracranial sarcoma: five cases report and review of literatures

 

Objective:

To investigate the clinicopathological characteristics and differential diagnosis of DICER1-mutant primary intracranial sarcoma.

 

Methods:

Five cases of DICER1-mutant primary intracranial sarcoma at Sanbo Brain Hospital, Capital Medical University during May 2013 to December 2024 were collected. The clinical and imaging data were retrieved. H&E and immunohistochemical staining, as well as next-generation sequencing, were performed. Additionally, a literature review was conducted. Results All five sarcomas were located in the supratentorial regions, with one case involving the basal ganglia, including three females and two males. The median age at diagnosis was 25 years (range: 8-33 years). Morphologically, they were characterized by malignant spindle cell sarcoma, with high mitotic activity and cytoplasmic eosinophilic globules. Myxoid degeneration, necrosis, and invasion into surrounding brain tissue were observed in some cases. Immunophenotypically, tumor cells showed diffuse expression of Vimentin and variable expression of myogenic marker (Desmin), with or without focal MyoD1 and/or Mogenin. Four cases of tumors exhibited diffuse strong positive expression of TLE1 and P53. Three cases displayed loss of nuclear ATRX expression in the tumor cells. Two cases showed mosaic loss of H3K27me3 expression in neoplastic cells. The Ki-67 proliferation index was significantly high (40-80%). Variable neuronal markers, such as Syn, NF, SOX2, MAP2, were expressed in all tumor samples. Genetically, all tumors samples harbored two DICER1 alterations, including a hotspot missense mutation in the RNase IIIb domain within exon 25 on one allele (p.E1813 or p.D1810) and a copy number deletion or mutation on the other allele (a germline mutation in one case). In addition, sarcomas showed alterations in TP53 (80%), ATRX (60%), and the mitogen-activated protein kinase pathway genes (60%). Finally, all five cases were diagnosed as DICER1-mutant primary intracranial sarcoma. All patients underwent craniotomy resulting in complete tumor resection. Patient 2 succumbed to the tumor after 3 months post-surgery due to rapid progression and tumor dissemination. Three patients accepted radiotherapy and chemotherapy following surgery, with progression-free survival times of 28, 48, and 50 months, respectively. One patient was lost to follow-up 3 months after surgery. Conclusion DICER1-mutant primary intracranial sarcoma, a newly defined tumor entity in the fifth edition of the WHO classification of central nervous system tumors, commonly occurs in children and young adults. High-grade malignant spindle cell sarcoma and eosinophilic cytoplasmic globules are their typical morphological features. Besides myogenic differentiation, these tumors may also exhibit neuronal differentiation. Confirming the pathological diagnosis relies on genetic testing for DICER1 pathogenic alterations or DNA methylation profiling.

 

Key words: DICER1; Intracranial; Sarcoma; DNA methylation; Eosinophilic globule

 

 

El Helali, Aya

The University of Hong Kong

Combination Talazoparib - carboplatin for recurrent high-grade glioma with DNA damage repair deficiency (DDRd) “TAC-GReD” trial

 

Background:

Recurrent high grade gliomas (HGG) present a significant challenge in neuro-oncology due to their poor prognosis and lack of effective standard treatment. Emerging evidence indicates deficiencies in the DNA damage repair (DDR) pathway in a significant proportion of gliomas, leading to innate or acquired resistance mechanisms and worse survival outcomes. Somatic alterations in the DDR pathway, including PTEN mutations and ""BRCA-ness"", have paved the way for targeted therapy utilizing poly (ADP-ribose) polymerase (PARP) inhibitors. The distinct mechanisms of action of PARP-trapping inhibitors, particularly in forming cytotoxic PARP-DNA complexes, have shown promise in personalized cancer treatment strategies.

 

Methods:

The TAC GReD trial is a prospective phase II, single-arm, open-label study designed to evaluate the efficacy and safety of the combination of talazoparib and carboplatin in patients with recurrent HGG harboring pathogenic mutations in the DDR pathway. The trial aimed to assess the 6-month progression-free survival (PFS-6) as the primary endpoint, with secondary endpoints including objective response rate (ORR), overall survival (OS), safety and tolerability, and health-related quality of life (HRQL) measures.

 

Results:

Thirty-three patients, with a median age of 55 and 70% male, were enrolled in the TAC GReD trial. The trial met the prespecified response criteria, with a PFS-6 survival rate of 25% (95% CI 13–48%), median progression-free survival of 3.4 months (95% CI 2.2–5.7%), and overall survival of 6.5 months (95% CI 4.5–9.8). The treatment combination was deemed safe, with the most common TRAEs being hematologic, and no treatment-related deaths were reported. The quality of life scores remained stable throughout the trial.

 

Conclusion:

The unique biomarker-stratified approach capitalizes on the strong biological rationale for utilizing PARP inhibitors in HGG, offering a potential avenue for innovative therapeutic strategies in the management of recurrent HGG.

 

 

Fang, Deyu

Northwestern University

A novel therapy with onco-targeting and immune-boosting dual efficacies in combating a broad spectrum of human cancers

 

Immunotherapies have revolutionized the treatment landscape for various solid tumors. However, only a subset of cancer patients benefit from the current checkpoint blockade therapies. Recent research underscores the impact of an immune-suppressive tumor microenvironment, marked by increased Treg infiltration and limited neoantigen presentation, as key factors driving resistance to immune checkpoint blockade. Furthermore, the integration of immune checkpoint inhibitors with standard chemotherapy, termed chemoimmunotherapy, has emerged as a cornerstone approach for several cancers. However, the immune-depleting toxicity of chemotherapeutic regimens often undermines chemoimmunotherapy efficacy. This highlights an urgent need for innovative precision therapies that effectively target both the tumor and bolster the immune response to overcome this resistance. Our laboratory has pioneered research on ubiquitin-specific peptidase 22 (USP22) over the past decade, establishing it as an oncogene (Molecular Cell, 2012). More recently, we demonstrated that USP22 promotes Treg-mediated immune suppression (Nature, 2020), identifying it as a dual-action target to both inhibit tumor growth and enhance antitumor immunity. Building on these insights, we developed the first USP22-specific small molecule inhibitor, which demonstrates both oncogenic targeting and immune-boosting activity with minimal toxicity across multiple solid tumor types (Science Advances, 2022). Our latest research further shows that USP22 enables tumors to evade immune surveillance by suppressing neoantigen presentation, providing a mechanistic basis for targeting USP22 to overcome immunotherapy resistance. With these insights, we have advanced to the development of a second-generation USP22-specific inhibitor with markedly improved antitumor efficacy. We are currently completing IND-enabling studies, with the ultimate goal of conducting the first-in-human Phase 1 clinical trial of this USP22-specific inhibitor in solid tumors, including glioblastoma.

 

 

Fong, Vernon

Hospital for Sick Children

Defining the involvement of the perivascular niche in brain tumor metastases

 

Medulloblastomas (MB) are the most prevalent malignant brain tumors in children, arising in the cerebellum or dorsal brainstem. These tumors are categorized into four distinct subgroups: WNT, SHH, Group 3, and Group 4, each characterized by unique gene expression profiles, metastatic behaviors, and recurrence rates. Importantly, metastatic progression is often associated with poor prognosis, impacting about 30% of patients at diagnosis. Despite extensive research on primary tumors, there are currently no approved treatments specifically targeting MB leptomeningeal metastases. Consequently, enhancing our understanding of metastatic MB is essential for developing targeted therapies.

 

The leptomeningeal niche, where metastatic MB cells reside, is comprised of a complex network of blood vessels supported by perivascular cells, including pericytes, smooth muscle cells, and fibroblasts. However, the specific adhesion molecules and signaling pathways that facilitate tumor cell colonization in this niche remain largely undefined. In this study, we hypothesize that the perivascular niche plays a critical role in the colonization and survival of metastatic cells within the leptomeninges.

 

To investigate this hypothesis, we utilized an innovative sLP-mCherry niche labeling system alongside single-cell RNA sequencing to spatially identify and analyze the perivascular cells surrounding metastatic MB tumor cells. Our analysis revealed upregulated genes linked to cholesterol export in these tumor-associated perivascular cells, indicating their role in facilitating metastatic cell colonization. To further explore this mechanism, we employed CRISPR and lentiviral shRNA constructs to knock down lipid receptors in MB cells, thereby reducing cholesterol uptake from the leptomeningeal microenvironment, which led to significant decrease in metastatic burden. Additionally, we observed that inhibiting perivascular recruitment to the tumor site—by decreasing chemokine secretion in MB cells—resulted in a reduced metastatic burden.

 

In summary, our study underscores the importance of the perivascular niche in metastatic medulloblastomas and provides novel insights into the molecular mechanisms driving tumor cell colonization within the leptomeninges. By exploring lipid transport mechanisms in the vascular microenvironment, we open new avenues for developing innovative treatments targeting MB metastases.

 

 

Giamas, Georgios

University of Sussex & Zhejiang Chinese Medical University

Developing an extracellular vesicles-based liquid biopsy biomarker signature of glioblastoma and comparison to whole genome sequencing plus tissue proteomics data using samples from the Genomics England Biobank

 

Background:

Despite significant advancements in the understanding of gliomagenesis, there remains no glioblastoma subtyping framework that is used in clinical practice. Additionally, diagnosis requires an invasive tissue biopsy, which limits the ability to perform longitudinal sampling resulting in a more limited understanding of the changes that occur in response to treatment and over time compared to other solid tumours.

Extracellular vesicles (EVs), small lipid membrane bound particles that contain various bioactive molecules including proteins, RNA and DNA, have been proposed as a potential liquid biopsy biomarker as their cargo has been demonstrated to reflect their cell of origin. Through the integration of plasma derived-EVs proteomics, transcriptomics and Raman spectroscopy signature, whole genome sequencing data, and tissue proteomics we aim to identify biologically and clinically relevant glioblastoma subtypes.

 

Methods:

Using 1mL glioblastoma plasma samples from the Genomics England biobank, and sex- and age-matched volunteer plasma samples, EVs were separated by size exclusion chromatography and characterised by protein quantification, nanoparticle tracking analysis, transmission electron microscopy and Western blot analysis to confirm EV enrichment. Differences in yield and particle size were assessed using an unpaired Welch’s t-test. Subsequently, analysis of the proteomic and transcriptomic cargo and label free Raman spectroscopy of the isolated EVs was investigated and bioinformatic analysis performed to identify a glioblastoma biomarker signature. Further comparisons with the whole genome sequencing data and tissue proteomics were investigated.

 

Results:

Plasma samples from glioblastoma patients from the Genomics England biobank have been analysed. Patients were predominantly male (63%) with a median age of 53. EVs have been separated and characterised from 57 glioblastoma and 49 healthy volunteer plasma samples. Following EV separation there was no difference in the mean protein yield (362.4 vs 244.4 μg protein, p=0.5860) or mean particle yield (1.28 x1011 vs 1.18 x1011 particles, p=0.5645) from 1mL plasma samples between glioblastoma patients and healthy volunteers. However, there was a significant difference in the mean (133.6 vs 114.9 nm, p<0.0001) and median (121.5 vs 103.4 nm, p<0.0001) particle size which is suggestive of altered EV biogenesis.

Multi-omic analysis of ~1000 identified EV protein species and EV micro-RNA cargo identified significant differences between glioblastoma and healthy volunteer plasma EV samples. Bioinformatic analysis, using a machine-learning algorithm, incorporated the identified multi-omic EV cargo plus Raman spectroscopic signature into a glioblastoma biomarker signature.

The developed EV biomarker signature will be added to clinicopathological, treatment and whole genome sequencing data to develop a comprehensive prognostic model. The identified signature will also be compared to tumour tissue-based mass-spectrometry proteomics and tissue spatial proteomics to investigate novel glioblastoma subtypes and the cellular source of the components of the identified biomarker signature.

 

Conclusion:

The development of an EV liquid biopsy signature for glioblastoma will have numerous applications, including a role in early diagnosis, development of a prognostic/predictive biomarker model, and patient monitoring during and following treatment. Additionally, novel biological insights related to intrinsic glioblastoma subtypes and tumour biology are likely to be identified.

 

 

Hassuneh, Osama

the University of Hong Kong

Developing Lipid Nanoparticles for Combinatory Chemo-Immunotherapy of Glioblastoma

 

Glioblastoma (GB) is an aggressive type of glioma, treated by a combination of surgery, radiotherapy and chemotherapy. However, its prognosis is still dismal. In recent decades, immunotherapy has revolutionised cancer treatment. Nonetheless, as monotherapy, immune modulation did not significantly improve treatment outcomes, as multiple clinical trials demonstrated disappointing results, specifically for GB patients. The GB tumour microenvironment (TME) is enriched with myeloid-derived suppressor cells, in addition to GB cells expressing surface proteins, such as CD47, which inhibit macrophage-mediated phagocytosis. Immunogenic cell death (ICD) is a specific apoptotic mechanism in which dying cells release stress molecules that trigger immune cell maturation and activate the adaptive immune response.

 

This research focuses on developing lipid nanoparticles for co-delivery of small interfering RNA (siCD47) to downregulate CD47, alongside an ICD inducer to boost the immune response within the microenvironment of GB.

 

Lipid nanoparticles (LNPs) have recently attracted attention as efficient carriers of nucleic acids (NA) for a wide range of applications, from COVID-19 vaccines to gene therapy agents providing a high encapsulation efficiency (EE) with minimal cytotoxicity. Here, LNPs co-encapsulating siCD47 and an ICD-inducer drug, including doxorubicin (DOX), mitoxantrone (MIT) or oxaliplatin (OXA), were prepared with a biocompatible size of (< 140nm) polydispersity index of (< 0.2) and a high %EE of siCD47 (> 90%). Drug encapsulation into LNPs was optimised via statistical analysis and mathematical models using the Design of Experiments (DOE) approach. In vitro cytotoxicity and ICD induction ability of the optimised LNPs were evaluated and compared with free drugs against brain cancer stem cells (BL6) and murine glioma cell line (GL261). BL6 cells were more sensitive to drug toxicity compared to GL261 cells. Drug toxicity was in the order of DOX>MIT>OXA. DOX and MIT encapsulation into LNPs showed reduced toxicity due to delayed drug release. Interestingly, DOX and OXA showed higher calreticulin (CALR) expression, an ICD marker, in the LNP form compared to free drugs. LNPsiCD47 reduced CD47 expression in BL6 (>70%) at doses > 10 nM. GL261 were more resilient to silencing in vitro.

 

LNPs encapsulating DOX and siCD47 were tested in an orthotopic GL261 mouse model following two intra-tumoral injections of 495 µg/Kg and 2300 µg/Kg for siCD47 and DOX, respectively. Groups receiving LNPsiCD47 and LNPsiCD47/Dox showed improved survival from 17 (untreated) to 20 and 19 days, respectively (sham Vs LNPsiCD47 or LNPsiCD47/Dox P < 0.05, LNPsiCD47 Vs LNPsiCD47/Dox P > 0.05).

 

In conclusion, we successfully developed LNP formulations with therapeutic potential in the GB mouse model. CD47 silencing shows promise in GB therapy. Although the combination with DOX showed no added benefit in this study, future work will explore the potential of OXA-LNP as a standalone or combination therapy.

 

 

Hung, Yuet Yi

The University of Hong Kong

Role of ENO1 in Glioblastoma Progression

 

Objective:

To investigate whether and how ENO1 contributes to glioblastoma (GBM) progression.

 

Method:

In vitro studies were conducted using human GBM cell lines, U87 and U251. Knockdown of ENO1 and inhibition of ENO1 using an ENO1 inhibitor, ENOBlock, were done on the cell lines to examine the role of ENO1 in GBM. The effects of ENO1 knockdown and inhibition were evaluated by western blot, immunofluorescence staining, cell viability and proliferation assay, cell cycle analysis by flow cytometry, and colony formation.

 

Result:

Treatment with ENOBlock or knockdown of ENO1 decreases the cell viability and cell proliferation of GBM, but increases its cell death and sensitivity to temozolomide (TMZ). Treatment with ENOBlock also decreases mitochondrial membrane stability in GBM. Knockdown of ENO1 reduces colony formation of GBM and decreases the expression of mitochondrial proteins.

 

Conclusion:

ENO1 contributes to the progression of GBM and can potentially be a therapeutic target. Coadministration of TMZ and agents inhibiting the ENO1 signalling pathway can potentially produce synergistic therapeutic effects.

 

 

Jiang, Yuanhao

University of Newcastle

Integrative Multi-Omics and Histopathological Imaging for Enhanced Subtyping and Clinical Management of Diffuse Midline Gliomas

 

Diffuse midline glioma (DMG) is an incurable and highly aggressive brain tumour with a poor prognosis and limited treatment options. Currently, early diagnosis DMG relies on the tumour’s location, histology-based morphology, and the staining for the hallmark loss of H3K27me3 which is inadequate for assigning molecularly distinct subtypes indictive of treatment recommendation. High-throughput profiling of DMG specimens, has identified genomic subtypes and potentially targetable recurring molecular features. However, the extended time required for these multi-omics assays to be performed limits immediate clinical application of potentially beneficial therapies, and hence interventions used later in the clinical journey of a patient have not yielded significant improvements in outcomes.

Here, we present a comprehensive overview of the molecular subgroups of DMG identified through advanced multi-omics approaches – including genomics, epigenetics – as well as imaging modalities based on histological sections. Systematically, published studies employing molecular profiling coupled with histopathological imaging were analysed employing a novel pipeline to integrate several distinct morphological feature analysis software’s we molecular signatures identifying tumour heterogeneity and patient outcomes.

 

Our findings indicate that integrated and matched multi-omics data and imaging offers a powerful strategy for identifying novel subgroups and actionable drivers, as has been demonstrated in the case of Medulloblastoma. Furthermore, the application of deep learning in imaging data analysis shows great promise in identifying specific features, enabling the diagnosis of molecular subgroups, such as IDH mutations in high-grade gliomas. This positions histopathological imaging as a rapid and informative alternative to traditional molecular diagnostics.

 

Our work emphasises the potential of combining multi-omics data with imaging biomarkers to enhance brain tumours subtyping, improve diagnostic accuracy and enable personalised treatment plans. The development of fine-tuned and robust deep learning models applied to matched multi-omics and histopathological imaging could revolutionise DMG management by facilitating real-time, precise subtyping and targeted therapeutic interventions tailored to each tumour entity. This integrated approach holds the promise of transforming clinical outcomes for patients affected by this devastating disease.

 

 

Khera, Sudeep

All India Institute of Medical Sciences, Jodhpur

Expression of SSTR-2 and HER-2-neu in Meningiomas and its correlation with clinico-pathological parameters

 

Meningiomas are one of the most common tumours of the central nervous system. Although commonly benign, atypical and anaplastic meningiomas also constitute part of the spectrum. Options for management are limited to surgery and radiotherapy. SSTR-2 has shown great promise as a target for therapy as well as intra-operative imaging of meningiomas. The expression of HER2/neu status has been substantial in various other malignancies for treatment purpose. However, it has shown variable results in meningiomas.

 

The present study was aimed to evaluate the expression of SSTR-2 and HER2/neu in meningiomas and to establish their correlation with various clinico-pathological parameters.

 

This was an ambispective observational cross-sectional study carried out in a tertiary care hospital in Western Rajasthan. This study included 120 cases of meningioma. On application of immunohistochemistry for SSTR-2 and HER2/neu on these cases, 75.83% cases showed positivity for SSTR-2 and none of the cases were positive for HER2/neu. SSTR-2 expression showed significant association with location of meningioma in brain and with histological subtypes. No significant association was found with age, gender, specific histo-morphological features and tumour grade.

 

None of the cases in this study were positive for HER2/neu. Previous studies also have showed a highly variable expression of HER2/neu in meningiomas. Hence, more studies are needed to investigate the role of HER2/neu in meningiomas as a diagnostic and prognostic marker as well as its role in targeted therapy.

 

Expression of SSTR-2 has been recommended by the 2021 WHO classification of CNS tumours as a desirable criteria for diagnosis of meningioma. In the present study,75.83% cases were positive for SSTR-2, however, a significant association with the grade and other clinical parameters could not be established. Since survival analysis was not performed, correlation with prognosis could not be assessed. Therefore, further studies are recommended to establish a significant clinico-pathological correlation as somatostatin receptor may have a significant role in confirmation of diagnosis of meningioma on imaging and in targeted therapy-based management in such cases.

 

 

Killilea, Joshua

Imperial College

From Innovation to Treatment: Bridging the Gap Between Engineering and Clinical Practice in Brain Tumor Therapy

 

Brain tumours are a group of malignancies characterised by high morbidity and mortality. The most common and aggressive primary brain tumour in adults is glioblastoma multiforme (GBM). For the past two decades, survival outcomes for GBM have remained stagnant with traditional avenues of surgery, chemotherapy, and radiotherapy being largely ineffective with the median survival time being 15 months. There is therefore an urgent need to develop improved treatment options for GBM patients. Developing novel pharmacological agents for GBM, however, represents a challenging and unrewarding prospect. As such, only 0.1% of novel anticancer agents for GBM progress from Phase 1 trials to registration. For other cancers, this number is 9%. Furthermore, developing novel pharmacological agents is hugely resource intensive from a time and financial perspective. Attempts to improve treatment outcomes in GBM, as well as other brain tumours, will therefore benefit from input from engineers to develop strategies to overcome current treatment limitation in GBM. For example, it is possible to develop stimuli-responsive vehicles for local drug delivery using ‘smart’ biomaterials, focussed ultrasound to transiently open the BBB and tumour treating fields. While such promising treatment options are currently being explored for GBM, critical to their successful translation will be effective interdisciplinary collaborations between engineers and clinicians. Such interdisciplinary collaboration is not always effective and can be challenging due to a significant gap in the mutual understanding of terminology as well as different ideas on which research questions are clinically important to address. We are conducting a global survey directed at clinicians and engineers to elucidate crucial insights regarding the quality of communication, comprehension of technical requirements and constraints, and the alignment of clinical needs with engineering solutions. Overall, the results demonstrate that clinicians are largely supportive and aware of the need for engineering advances to address challenging questions in cancer research, with all respondents agreeing with the statement that engineering input will be critical in addressing questions in cancer research. Furthermore, useful strategies for more effective inter-disciplinary collaborations are presented. Common strategies suggested included the need for a ‘common language’ between clinicians and engineers as well as shared reading material to help create shared research goals. Another key point was that clinicians need to continue to network with engineers and provide succinct summaries for them on current clinical challenges to set the scene for where engineers can help. To help facilitate the translation of such developed technologies, clinicians also think it would be useful to attend their clinical practice to see better the clinical workflow to ensure that engineers design something that is not also effective but clinically feasible. This survey highlights the imperative for a standardized framework to enhance engineer-clinician collaboration, thereby maximising the clinical impact of technological advancements in the treatment of brain tumours.

 

 

Lan, Yanjie

Beijing Tiantan Hospital, Capital Medical University

SYHA1813, a dual-target inhibitor of VEGFR and CSF1R, exerts antitumor activity in meningioma in vitro and in vivo

 

Meningioma is common tumors of the central nervous system, arise from the dura mater and the outer layer of the arachnoid. Malignant meningiomas are rare subtype, but they are generally more aggressive with high recurrence rate. Malignant meningiomas patients still lack effective therapy currently. Therefore, developing a novel regent is urgently needed. SYHA1813 is a novel dual-target inhibitor, which targets CSF1R and VEGFR. Our previous study suggested SYHA1813 exhibits potent preclinical antitumor effects in glioblastoma. It’s also reported that SYHA1813 has potentiality in treating B-cell lymphoma. However, the actual functional role of SYHA1813 in meningiomas remains unknown. Herein, we investigated the tumor inhibition effect of SYHA1813 on meningioma cells in vitro and in vivo.

 

To explore the cell toxicity of SYHA1813 in meningioma cells in vitro, we evaluated the anti-proliferation effect of SYAH1813 on two human meningioma cell lines. CCK8 assay results showed that SYHA1813 reduced the cell viability of IOMM-Lee and CH157 cells in a dose-dependent manner. Consistent with CCK8 assay, EdU assays showed SYHA1813 treatment resulted in a marked decrease in the fraction of EdU-positive cells. In addition, SYHA1813 suppressed colony formation of IOMM-Lee and CH157 cells, indicating that SYHA1813 could affect the long-term survival of meningioma cells. Wound healing assay and trans-well cell invasion assay revealed that SYHA1813 remarkably inhibited the migration and invasion of IOMM-Lee and CH157 cells. These results demonstrated SYHA1813 exhibited significant anti-proliferation activity in meningioma cells.

 

To determine whether the inhibitory effects of SYHA1813 on proliferation were due to cell cycle arrest or apoptosis. Meningioma cells were treated with different dose of SYHA1813 for 48h and analyzed the cell cycle and apoptosis by flow cytometry. We found SYHA1813 arrested cell cycle at G2/M phase and promoted cell apoptosis. Interestingly, enlarged and flattened IOMM-lee and CH157 cells following exposure to SYHA1813, which appealed us to explore whether SYAH1813 could induce cell senescence. SA-β-Gal staining assays showed that the augmentation of senescent cells, indicating that SYHA1813 could promote cell senescence in meningioma cells. Taken together, these findings suggested that SYHA183 treatment could block cell cycle, promote cell apoptosis and senescence, which leads to an inhibition of the proliferation in meningioma cells in vitro.

 

To further evaluate the functional role of SYHA1813 in meningioma in vivo, IOMM-Lee cells were inoculated into balb/c nude mice to monitor subcutaneous xenograft mouse model. We found tumor volume and weight of mice orally administered with SYHA1813 was effectively reduced compared to mice in vehicle group. Tumor cell proliferation was assessed by Ki67 immunohistochemistry (IHC) staining. Less Ki67-positive cells were observed in tumors from mice treated with SYHA1813. Collectively, consistent with in vitro studies, these results explained the role of SYHA1813 in inhibiting meningioma cell growth in vivo.

 

In summary, our study revealed the antitumorigenic role of SYHA1813 in meningioma cells in vitro and in vivo, proving the great potential of SYHA1813 in the treatment of malignant meningioma.

 

 

Lv, Jing

Beijing Normal University

Loss of temporally regulated differentiation of neural stem cells in IDH-wildtype glioblastoma

 

Background:

Isocitrate dehydrogenase (IDH)-wildtype glioblastoma (GBM) is highly heterogeneous and aggressive brain tumor in adults. Though understanding of molecular pathogenesis of IDH-wildtype GBM has been advanced, patients´ survival outcomes have not been significantly improved. Mapping GBM into neural lineages and differentiation stages may uncover hitherto inadequately characterized therapeutic targets. Based on the expression profile of the EGFR or PDGFRA co-expression module (the EM and PM, respectively), we proposed an EM/PM classification scheme and assigned adult gliomas into EM or PM molecular subtypes that are distinct in clinical phenotypes, genomic alterations, and resemblance to neural lineages. EM subtype gliomas correspond to IDH-wildtype GBM; PM subtype gliomas contain IDH-mutant astrocytoma and oligodendroglioma. EM and PM subtypes are stable during progression of individual patients and across the regions and single cells of individual gliomas. Glioma progression is not driven by a switch of molecular subtypes but by genomic instability and microenvironment. Here we mapped the aberrant neural developmental program of IDH-wildtype GBM.

 

Methods:

Using bulk transcriptomes, single-cell transcriptomes and proteomes, GBMs were mapped into neural lineages. Expression patterns of signatures of neural stem cells (NSC) and neural progenitor cells (NPC) in individual IDH-wildtype GBM cells were assessed using single-cell RNAseq analysis and RNAscope on clinical samples. We assessed the expression patterns of key regulators in neuronal and oligodendrocyte lineage in GBM transcriptomes. Conversely, we also assessed the expression pattern of IDH-wildtype GBM-enriched genes in the transcriptomes of brain development.

 

Results:

Signatures of NSC and neural NPC are enriched in the transcriptomes and proteomes of IDH-wildtype GBMs. In contrast, signatures of neuronal and oligodendrocyte lineage are significantly down-regulated in IDH-wildtype GBMs. IDH-wildtype GBM cells are correlated to NSCs and NPCs. Canonical markers of NSC and NPC are enriched in distinct cell populations of IDH-wildtype GBM cells. Regulators of fate decision and development of neuronal and oligodendrocyte lineages are downregulated in IDH-wildtype GBMs. Expression patterns of IDH-wildtype GBM-enriched genes during human brain development indicate loss of temporally regulated differentiation of neural stem cells. Expression pattern of those temporally dysregulated genes in mouse brain development indicates precocious astrocyte genesis in IDH-wildtype gliomas, which coincides with abnormal activation of Notch and STAT signaling.

 

Conclusions:

Our studies show that IDH-wildtype GBMs resemble NSC and NPC and are blocked from differentiation into mature neuron or oligodendrocyte. Abnormal activation of Notch and STAT signaling in NSC may induce precocious astrocyte genesis and inhibit neuronal and oligodendrocyte differentiation, eventually cause IDH-wildtype GBM genesis. These findings indicate that restoring differentiation could be an alternative therapeutic approach for IDH-wildtype GBM.

 

Key words: IDH-wildtype GBM; EM/PM classification; brain development; neural differentiation

 

 

Maas, Sybren

Leiden University Medical Center

IDH-mutant astrocytomas with primitive neuronal component have a distinct methylation profile and a higher risk of leptomeningeal spread

 

IDH-mutant astrocytomas are diffuse brain tumours that are defined by characteristic mutations in IDH1 or IDH2 and do not have a 1p/19q co-deletion. The established grading criteria include histological features of brisk mitotic activity (grade 3), necrosis or microvascular proliferation (grade 4). In addition, a homozygous deletion of the CDKN2A/B locus is implemented as a molecular marker for grade 4 IDH-mutant astrocytomas. We describe a subgroup of IDH-mutant astrocytomas defined by primitive neuronal morphology based on histology and a distinct DNA methylation profile (n=51, ASTRO PNC). Misinterpretation as carcinoma metastasis was common, since GFAP expression was absent in the primitive neuronal component, whereas TTF1 expression was detected in n=14/18 (78%) of cases based on immunohistochemistry. Targeted DNA panel sequencing revealed enrichment for alterations in RB1 (n= 19/51, 37%) and MYCN (n= 11/51, 22%) besides the astrocytoma-characteristic mutations in IDH1, TP53 and ATRX. Homozygous CDKN2A/B deletions (n= 1/51, 2%) and CDK4 amplifications (n= 3/51, 6%) were relatively rare events in our cohort.

The clinical (n=31) and survival (n=23) data we collected indicate a biological behaviour similar to WHO grade 4 IDH-mutant astrocytomas (p= 0,2446) with an increased risk for leptomeningeal (n=7) and extra-CNS (n=2) spread. Taken together, ASTRO PNC is defined by a distinct molecular and histological appearance that can mimic metastatic disease, featuring an aggressive clinical course.

 

 

Mahdi, Haridini Intan Setiawati

Indonesian National Cancer Center-Dharmais Cancer Hospital, Jakarta, Indonesia

Clinical Characteristics of Pediatric Brain Tumors : A Single Institution Experience in NCC Dharmais Cancer Hospital, Indonesia

Background:

Pediatric brain tumors are the most common solid tumors among children aged 0–14 years and remain a leading cause of childhood cancer mortality. The clinical characteristics of these tumors are diverse, encompassing variations in developmental origin, genomic profiles, therapeutic approaches, and outcomes. While advancements in surgical techniques, neuro-oncology, neuroradiology, and radiation oncology have significantly improved survival rates, certain pediatric brain tumors continue to have poor prognoses. Understanding these clinical characteristics is essential for enhancing diagnosis, management, and prognosis of pediatric brain tumors.

 

Objective:

To describe the clinical characteristics of pediatric brain tumor patients treated at the National Cancer Center, Dharmais Cancer Hospital, Jakarta, Indonesia.

 

Methods:

This cross-sectional descriptive study analyzed medical records from Dharmais Cancer Hospital between July 2021 and June 2024. Using consecutive sampling, 79 pediatric brain tumor cases were reviewed.

 

Results:

Among the 79 cases that met the study criteria, most patients were male (64.5%) and were predominantly diagnosed at ages 0-9 years (56.9%). Headache was the most common presenting symptom, leading to hospital admission (39.2%). Tumor location was frequently found in the cerebellum and fourth ventricle (36.7%), with gliomas (30.3%), medulloblastomas (26.5%), and ependymomas (15.1%) identified as the most common histological types. The majority of patients (37.9%) received a full combination of therapies, including surgery, chemotherapy, and radiation. Despite comprehensive treatment, outcomes were sobering, with 46.8% of patients succumbing to the disease, 43.0% surviving, and 10.1% lost to follow-up. The median time to diagnosis was 3 months, and the median treatment initiation time was 1 month.

 

Conclusion:

This study highlights a high prevalence of brain tumors among young pediatric males presenting with headaches as a primary symptom. Tumors predominantly affected the cerebellum and fourth ventricle, with glioma being the most frequent histological type. While multi-modal treatments are widely utilized, the high mortality rate underscores an urgent need for improved diagnostic and treatment strategies. Enhanced understanding of these clinical characteristics can guide future research and therapeutic innovations, ultimately aiming to improve survival and quality of life for pediatric brain tumor patients in Indonesia.

 

 

Mai, Hoang Vu

Tam Anh General Hospital

Evaluation of results in intracranial gliomas surgery by using synaptive Modus V robotic system at Tam Anh General Hospital Ho Chi Minh City in Vietnam

 

Background:

Glioma is one of the common primary intra-axial tumors in CNS. The age-standardized incidence rate of gliomas was 4.7 per 100,000 person-years. Glioma tumor surgery has two goals: removed tumor and preserved neurological functions. The Modus V Synaptive robot includes an exoscope system with the robotic arm that integrates a modern navigation system in neurosurgery. This navigation system can regenerate the nerve fiber bundles (DTI) during operation, allowing the neurosurgeons to access the lesions without affecting the nerve conduction bundles for surgery.

 

Objective:

To evaluate the results of gliomas surgery using the Modus V Synaptive robot system at Tam Anh General Hospital HCMC.

 

Methods:

A prospective descriptive study of a series of cases with a diagnosis of gliomas operated by the Modus V Synaptive robot system at Tam Anh General Hospital HCMC from January 2023 to July 2024.

 

Results:

Among 43 cases were operated by Modus V Synaptive Robot system at Tam Anh General Hospital HCMC from 01/2023 - 06/2024. Median age’s 43.6 years. The ratio of male: female is 1:1. Common symtomes are headache (93.0%), followed by nausea (81.4%), weakness (65.1%), seizure (23.3%), reduce vision (18.6%), ataxia (16.3%). Tumor locations varied, in supratentorial with frontal (23.3%), temporal (13.9%), ventricular and thalamic involvement 9.3% of cases, followed by basal ganglia and caudate nuclei (7.0%), parietal (7.0%), occipital (7.0%), optic pathway (7.0%), hippocampus (4.7%), and pineal region (4.7%) in- volvement. For infratentorial, the tumors were located in cerebellar hemisphere, vermis and brainstem (9.3%, 4.7% and 2.3% respectively). On the preoperative MRI- DTI, the change of tracts with tumors were divided into 4 types: normal (14%), displacement or edematous (48.8%), infiltration (27.9%), discruption or destroyed (9.3%). Transsulcal parafascicular using tubular retractors (Vycor, NICO) was performed in 21 cases (48.8%). The histopathology of tumors were classified with grade I involvement 15 cases (34.9%), followed be grade II (32.6%), grade III (14.0%) and grade IV (18.5%). Gross total resection (GTR) was achieved in (60.5%) of cases, near-total resection (NTR) in (23.2%), and subtotal resection/partial resection (STR/PR) in (16.3%). The recovery of neurological functions significantly different between GTR and STR/NTR/PR (p<0.0001). Postoperative complications included hemiparesis or weakness (11.6%), CFS leak (9.3%), infections (7.0%), visual deficits (4.7%), seizure (4.7%). The GOS score at discharge from the hospital was at 5 : 34 cases (79%), followed by GOS3-4 with 6 cases (14.0%), GOS1-2 with 3 cases (7.0%).

 

Conclusion:

The neurotransmission recovery after surgery is related to the patient's discharge GOS score. The GTR has been showed good results and clinical patient improved. Robotic surgery achieves the positive effect in brain tumor surgery when nerve fiber bundles are preserved as much as possible.

 

 

Miceli, Giovanni Carlo

Imperial College London

Overcoming the Blood-Brain Barrier: A Polymeric Bioelectronic Implant for Targeted, Voltage-Driven Drug Delivery

 

Brain tumours are characterised by high morbidity and mortality owing to their localisation and often locally invasive growth. Glioblastoma multiforme (GBM) represents the most common malignant tumour in the central nervous system (CNS), accounting for 14.2% of all tumours and 50.9% of all malignant brain tumours. Despite advances in neurosurgery, chemotherapy, and radiotherapy, it remains one of the most treatment-resistant CNS malignancies, and the tumour inevitably recurs. Survival rates for GBM have remained unchanged since the introduction of the Stupp protocol almost 20 years ago. The prognosis is only 15 months, with less than 5% of patients surviving beyond 5 years even with aggressive treatment. Attempts to design pharmacological agents capable of treating glioblastoma are hampered by tumour heterogeneity, immune privilege of the central nervous system and the blood-brain barrier. The primary chemotherapeutic agent used to treat these tumours is temozolomide due to its ability to cross the blood-brain barrier (BBB). The BBB is a pivotal obstacle to GBM treatment since it regulates the entry of molecules into the brain. Local drug delivery platforms offer the ability to get drugs to target cells while minimising systemic delivery and adverse events associated with treatment. Some of the recent devices developed for tissue localised clinical administration of drugs have been focused on using pressure to inject drug solutions into the diseased tissue area. This method relies on pressure differentials causing increased intracranial pressure and drug reflux. To address this challenge, we have developed a polymeric bioelectronic implant to control the drug concentration in the tumour microenvironment by leveraging the voltage application. This strategy aims to overcome chemoresistance and unnecessary exposure to healthy tissues by circumventing the BBB for high-precision chemotherapeutic targeted drug delivery. This bioelectronic implant consists of a polyurethane matrix with a conductive polymeric filler for voltage-controlled dry release of drug molecules. The material enables the encapsulation and stabilising of small molecule drugs and on-demand electrically controlled offloading of the drugs. Proof-of-concept has been demonstrated using anionic and cationic dyes, demonstrating that their release can be enhanced at repulsive and inhibited at attractive voltages. This device has the potential to load several charged chemotherapeutics and precisely release them within the tumour. Preliminary experiments have been performed using doxorubicin, paclitaxel and cisplatin, using voltages well within the range of safe human levels, demonstrating that we are indeed able to control the release of anionic and cationic chemotherapeutic drugs. Given the tumour heterogeneity of GBM, this feature is crucial to target multiple tumourigenic molecular pathways to reduce chemoresistance and recurrence. The implant uses electrophoretic release to control the unloading of charged chemotherapeutic agents and can improve tissue penetration over passive local delivery approaches. Moreover, electrical stimulation through the drug-releasing electrode may allow modulation of proliferative potential and drug sensitivity. Multimodal treatment through locally targeted chemotherapy and electrical modulation of cancer cell biology may yield unexpected synergies that can finally make a significant step towards curing patients.

 

 

Nguyen, Nhu

Children's Hospital 2

Clinical Characteristics and Treatment Outcomes of Intracranial Germ Cell Tumor at Children's Hospital 2 in the South of Vietnam

 

Objective:

Intracranial germ cell tumors (IGCTs) are an uncommon neoplasm that predominantly impacts children and young adults. Pediatric IGCTs represent a significant challenge in low-income nations due to restricted access to specialized care and treatment resources may be limited. This study aims to provide essential information regarding Characteristics, Clinical Features, Treatment Approaches, and Outcomes of Intracranial Germ Cell Tumor at Children's Hospital 2.

 

Method:

A retrospective analysis of twenty-seven pediatric patients diagnosed IGCTs at Children’s Hospital 2 in Ho Chi Minh City, Vietnam, from May 2020 to November 2022, with follow-up extending until May 2023.

 

Results:

The study found a male-to-female ratio of 4.4:1 among participants, with a mean age of diagnosis at ten years. The median interval from symptom start to diagnosis was two months. The most commonly reported symptoms were headache and nausea or vomiting, impacting approximately 52% of the patients. Subsequently, localized neurological deficits manifested in around 33% of cases, whereas endocrine disturbances were observed in 22% of subjects. Magnetic resonance imaging indicated that the most common locations of tumors were the pineal and pituitary glands, accounting for 44% and 33% of cases, respectively. No cases of metastasis were detected at the time of diagnosis. The number of patients with increased AFP and beta HCG in blood and cerebrospinal fluid and without increased AFP and beta HCG was almost equal, at 49% and 51%, respectively. Endocrine disorders, especially hypopituitarism, were common in patients with pituitary gland tumors, impacting 78% of this group. Following the guidelines and protocols of the Société Internationale d'Oncologie Pédiatrique Central Nervous System Germ Cell Tumor II (SIOP CNS GCT II), the majority of patients received surgical intervention, with nine patients undergoing partial resection (33%) and six patients undergoing total resection (23%). In addition to surgery, all patients received chemotherapy, and twenty patients (74%) experienced radiotherapy as part of a multimodal treatment approach. The 12-month survival rate was 76%, with a median survival duration of 23 months. Notably, no tumor recurrences were observed by the end of the study period.

 

Conclusion:

This study offers significant insights into the clinical presentation, treatment, and outcomes of pediatric IGCTs at Children's Hospital 2, Vietnam. Despite constrained resources in a low-income environment, the findings have the potential for favorable short-term outcomes using a multimodal treatment strategy comprising surgery, chemotherapy, and radiotherapy.

 

 

Pitchaimani, Arunkumar

Vellore Institute of Technology (VIT)

Mitochondrial transplantation: an organelle-mediated drug delivery system for potential glioblastoma multiforme therapy

 

Organelle-mediated drug delivery is a novel bioengineering approach exploiting the usage of the mammalian cellular organelle to deliver therapeutic drugs/genes for various biomedical applications. Among them, mitochondria(MT), a powerhouse of the cell play a crucial role in activating the apoptosis of the mammalian cells. With the advent of MT transplantation technology, MT has been exploited to overcome the current clinical challenges in many mitochondrial diseases. However, in cancer therapy, most of the drugs/therapeutics are more focused on only targeting MT in cancer cells. Based on the clinical layouts and the biopotential of MT-transplantation, we hypothesized that MT-transplantation in the presence of the anticancer drug will enhance apoptotic signatures in tumor cells. The utilization of MT as a drug delivery vehicle has not been explored so far. In this study, we investigated the drug-loading strategies of functional MT and studied its anti-cancer efficacy against glioblastoma multiform model in mice models. For this, a functional MT from Mesenchymal stromal cells was isolated and characterized for its stability through size, morphology, membrane potential, ATP production, etc. Further, MT was loaded with the model chemotherapeutic drug, doxorubicin. Using bioengineering approaches, we developed novel strategies to load higher concentrations of drug molecules into mitochondria without affecting their viability and membrane integrity. The novel strategies include (1) 37C incubation; (2) Saponin treatment (3) Freeze-Thaw (4) Electroporation. With DOX-loaded-MT, we investigated its in vitro anti-tumor potential in U87MG cell lines and also evaluated its PK-PD parameters, brain accumulation potential, and in vivo anti-glioblastoma efficacy in preclinical mice models. The results showed that the functional mitochondria were successfully isolated from the MSC cells and characterized for their size, yield, and protein concentrations. With our experimental setup, we also found the exogenous cell-free mitochondria can also be taken up by the cell through the process of endocytosis. Further results show enhanced cellular uptake and intracellular distribution in U87MG after 6h incubation. It may be envisioned that supplying energy to the cancerous cells in the form of MT transplantation can worsen the condition, but our preliminary in vitro results showed that donor mitochondria induce significant apoptosis in a U87MG cell. A dose-dependent cytotoxicity and significant apoptosis induction were observed in U87MG cells after 24h incubation. In vivo, biodistribution shows enhanced brain accumulation through intranasal administration compared to intravenous. The in vivo-antitumor efficacy of the DOX-loaded-MT study is ongoing, and the results will be compiled soon. Thus, the bioengineered mitochondria with DOX payload will play a huge role in future glioblastoma research with enhanced bioaccumulation in the brain for advanced glioblastoma therapy and can be used as an effective therapeutic tool for personalized GBM therapy.

 

 

Plate, Karl H.

Goethe University

Identification and functional phenotyping of lymphoid aggregations in brain metastases

 

Lymphoid aggregations (LA), such as tertiary lymphoid structures (TLS), have been shown to predict overall survival and response to immune checkpoint therapy in non-CNS cancers. Data on tumors of the CNS, however, are sparse. Here, we applied CD20 bright-field immunohistochemistry as a screening tool to identify LA in a retrospective cohort of 461 FFPE-brain metastasis samples, derived from the archives of the University Cancer Center (UCT) Frankfurt, Germany, between 2015 and 2022. Stereotaxic biopsies as well as spinal/vertebral metastases were excluded from the study. LA-positive tumors were further analyzed by multispectral whole slide TSA-immunofluorescence (IF) using antibodies for von Willebrand factor, CD3, CD20, CD163, Lamp3, pan-cytokerarins, and DAPI as counterstain. TSL-positive tumors were further characterized by whole exome sequencing, bulk RNA sequencing, spatial transcriptomics and customized highplex-sequential IF panels that included > 40 antibodies for the analysis of tumor immunology and microenvironment.


TLS were observed in 38% of brain metastases and were most common in lung to brain metastasis (46%). Our preliminary data revealed a survival benefit in patients with TLS+ brain metastasis. By RNAseq, spatial transcriptomics and highplex-IF, three different subtypes of TLS were identified, based on their cellular and molecular compositions. Interestingly, most of the TLS identified lacked the structural organization reminiscent of a germinal center, suggesting an abrogated immune response. Further analysis of the TLS subtypes is being conducted to investigate their biological significance and clinical relevance in patients’ response to therapy.

 

 

Puri, Sameer

All India Institute of Medical Sciences (AIIMS), New Delhi

SPECC1L-NTRK2 Fusion in High-Grade Glioma in a Young Adult: A Rare Case Study and Diagnostic Implications

 

Background:

Neurotrophic tyrosine receptor kinase (NTRK) gene fusions are identified in less than 2% of gliomas, with a prevalence of less than 1% across all tumors. NTRK encodes for tropomyosin receptor kinase (TRK), which is highly expressed in neural tissue and regulates neuronal developmental pathways through the MAPK, PI3K/AKT and PLC-γ signalling cascades. NTRK fusions, particularly SPECC1L-NTRK2 fusions, have been identified across various pediatric and adult tumors, including gliomas. The clinical identification of NTRK-fusion is important due to the availability and clinical efficacy of the newly approved TRK inhibitors in the treatment of NTRK-fusion tumors and shows good prognosis irrespective of the tumor type, histology or grade. The detection of these fusions via whole-genome sequencing (WGS) and whole transcriptome RNA sequencing (RNAseq) has opened new avenues for precision medicine, offering targeted therapies in cases where standard treatments may be ineffective.

 

Case History:

We report the case of a 26 year old male presenting with progressive headaches and neurological deficits. MRI revealed a mass in the left insular region with aggressive features including irregular thick margins, central necrosis, marked mass effect, hemorrhage, intense irregular heterogeneous enhancement of margins with nodular components and elevated perfusion.

 

Observations:

Surgical resection was done and tumor tissue send for histopathological examination. Sections examined showed features of a high grade glioma with endothelial proliferation and areas of necrosis. On immunohistochemistry, tumor cells were immunonegative for IDH1 R132H and p53 while ATRX was retained. MIB-1 labelling index was 10-15% in highest proliferating area. Thus, a histopathological diagnosis of glioblastoma, WHO Grade 4 was given initially.Further molecular profiling confirmed the presence of a SPECC1L-NTRK2 fusion, refining the diagnosis to NTRK fused high grade glioma. Despite the aggressive tumor characteristics, the patient showed stable disease 20 months post-surgery, without evidence of progression.

 

Conclusion:

This case highlights the significance of identifying SPECC1L-NTRK2 fusions in gliomas, as it opens the door for targeted and highly effective therapies like TRK inhibitors. With the increasing detection of NTRK fusions in various cancers, incorporating advanced molecular techniques such as WGS and RNAseq can significantly impact clinical outcomes, offering more personalized treatment options.

 

 

Ratna, Sylvanie

Dharmais NCC Hospital

Serial cases : primary neuroendocrine carcinoma (NEC) of the brain: diagnostic approach and treatment

 

Introduction:

Primary neuroendocrine carcinoma (NEC) of the brain is a rare case with challenging diagnostic and treatment approach. Many cases report distinct characteristics of the patient, treatment approach and prognosis. Our case report two different group of age with identical immunohistochemistry result of NEC.

 

Case Report:

First case is a 70-year old patient who visited our outpatient clinic with progressive right-sided weakness three months before admission. He also has focal to generalized with impaired awareness seizure. Brain MRI showed solitary brain lesion within left parietal lobe. Patient underwent craniotomy. The tissue immunohistochemistry (IHC) was positive for cytokeratin (AE1/3), synaptophysin, chromogranin, CD56, and negative GFAP. CT thorax, abdomen, and pelvis showed no other lesion. Surgery was followed by radiation therapy, 54 Gy in 30 fractions with planned brain MRI 12 weeks after radiotherapy. We preferred not to give chemotherapy because of his age.

 

Second case is a 21-year old woman with increased intracranial pressure symptoms and seizure over one month ago. Brain MRI showed solitary lesion within left frontal lobe. Tumor removal was done and histopathology accorded with malignant tumors with papillary pattern. IHC was positive for EMA, AE1/3, CD56, NSE, and negative for synaptophysin, chromrogranin, GFAP, vimentin, AFP, GATA 3, thyroglobulin, TTF-1, S100. There was no evidence of another extracranial lesion by associating PET/CTs. A total dose of 54 Gy in 30 fractions was successfully done two months after surgery. Chemotherapy was not performed because of poor patient compliance. Eight months later the tumor became progressive, then tumor removal followed by radiation therapy was repeated without chemotherapy. One year later, the tumor progressed to double in size.

 

Discussion:

Neuroendocrine carcinoma (NEC) of the brain is a rare case categorized as functional and non-functional. Gender and age specific has not been reported, so do our cases that show different patients characteristics and histopathology. Both of our patient have identical IHC, positive AE1/3 and C56 which can be found in NEC. Tumor removal and radiation therapy is the main treatment. Chemotherapy with cisplatin and etoposide is recommended for high grade NEC. We suggest to consider patient performance status, residual tumor and Ki67-positive cell, otherwise observe until progression occurs. First patient’s Karnofsky Performance Status (KPS) is 80 and has Ki67-positive cell >75%. Second patient’s KPS is 70 and has Ki67-positive cell >10% but with poor treatment compliance. We decided to suspend chemotherapy for first patient yet offer it to the second patient.

 

Conclusion:

Requirement of tumor removal and radiation therapy of primary brain NEC is an evidence-based treatment. Chemotherapy should be considered for high risk of recurrence cases. Prognosis differs due to different patient characteristics.

 

 

Shahi, Mehdi H.

Aligarh Muslim University

Potential novel molecule ARSH-Q for the treatement of most malignant brain tumor glioblastoma

 

Glioblastoma is still an aggressive brain tumor and causes high mortality, despite advanced neurosurgery and chemoradiation therapy. The relapses are common, and the main culprit is glioma-initiating stem cells (GISCs). Targeting these stem cells is crucial for the patient's survival and enhancement. Our lab screened multiple compounds to better target stem cells. Fortunately, we hit a molecule ARSH-Q which has the potential to kill glioma cells efficiently and regulate the reduction of stem cell markers BMI1, NANOG, OCT4, SHH-GLI1 cell signaling pathway expression and inhibited the growth of glioblastoma in vitro study. In addition, this novel molecular/compound also has a very low LD50 compared to the most common drug Temozolomide for glioblastoma growth inhibition. This potential compound may be used as a therapeutic target for the treatment of brain tumor glioblastoma.

 

 

Tang, Wanjun

The University of Hong Kong

Impaired degradation of IDH1 through Chaperone-mediated autophagy promotes glioblastoma cell cycle progression

 

Chaperone-mediated autophagy (CMA) selectively targets individual proteins containing KFERQ-like motifs for lysosomal degradation, primarily facilitated by the chaperone heat shock cognate protein of 71 kDa (HSC70) and the receptor lysosome-associated membrane protein type 2A (LAMP2A). While several CMA substrates have been identified in various cancers, the specific roles and substrates of CMA in glioblastoma (GBM) remain poorly understood. Notably, isocitrate dehydrogenase 1 (IDH1), a key metabolic enzyme and prominent GBM marker, has been identified as a CMA substrate in mouse embryonic stem cells. In this study, we investigated the role of CMA in GBM and its impact on IDH1 turnover. Our results demonstrated that inhibiting CMA through LAMP2A knockdown significantly promoted GBM cell growth both in vitro and in vivo, while LAMP2A overexpression suppressed cell proliferation. CMA inhibition disrupted the RB1 pathway and enhanced cell cycle progression, as indicated by increased levels of positive regulators such as Cyclin D1 and CDK4. Additionally, IDH1, which contains a conserved CMA motif, accumulated following LAMP2A knockdown or lysosomal inhibition and interacted with HSC70 in GBM cells. Functionally, suppressing IDH1 reversed the tumor-promoting effects of CMA inhibition and restored the dysregulated RB1 pathway, while IDH1 overexpression promoted cell cycle progression. Mechanistically, we showed that the metabolic product of IDH1, -ketoglutarate, partially stimulated Cyclin D1 expression. In conclusion, our findings suggest that CMA is essential for suppressing GBM progression by facilitating IDH1 degradation and inhibiting cell cycle progression.

 

 

Tang, Wanjun

The University of Hong Kong

IDH1 persistence due to compromised chaperone-mediated autophagy accelerates cell cycle progression in glioblastoma

 

Chaperone-mediated autophagy (CMA) is a selective protein degradation pathway crucial for cellular homeostasis. However, its role in glioblastoma, a highly aggressive brain tumor, is not well understood. In this study, we elucidate a tumor-suppressive role of CMA in glioblastoma through the degradation of isocitrate dehydrogenase 1 (IDH1), a pivotal metabolic enzyme and prominent molecular marker frequently upregulated in glioblastoma. Inhibiting CMA through knockdown of its receptor, lysosome-associated membrane protein type 2A (LAMP2A), significantly enhances glioblastoma cell growth both in vitro and in vivo, associated with dysregulation of the RB1 cell cycle pathway. We reveal that IDH1 possesses a conserved CMA targeting motif and interacts with the CMA chaperone heat shock cognate 71 kDa protein (HSC70). Intriguingly, knockdown of LAMP2A or lysosomal inhibition leads to IDH1 accumulation, suggesting that CMA deficiency promotes tumor growth by scavenging IDH1 protein. Consistent with these findings, IDH1 suppression counteracts the tumor-promoting effects of CMA inhibition and restores cell cycle regulation in CMA-deficient cells. In contrast, upregulation of IDH1 fuels cell cycle progression, partly mediated through its metabolic product, -ketoglutarate (-KG), which stimulates Cyclin D1 expression. These findings reveal a previously unrecognized role of CMA in scavenging IDH1 and regulating cell cycle progression to suppress glioblastoma cell growth.

 

 

Tang, Wenshu

Hong Kong Genome Institute

Single-cell long-read sequencing uncovers novel RNA isoforms and splicing variants in glioblastoma

 

Glioblastoma is a highly aggressive brain cancer with limited effective treatment options, driven by complex cellular heterogeneity. Alternative splicing plays a critical role in cellular differentiation and function, potentially driving tumor aggressiveness and treatment responsiveness. Notable examples include constitutive EGFR activation due to EGFRvIII variants and exon skipping in MET, which affects sensitivity to MET inhibitors. While short-read single-cell RNA sequencing (scRNA-seq) is commonly used to profile cellular populations, it often lacks the capacity to fully capture complete RNA isoforms. Long-read bulk RNA sequencing identifies isoforms comprehensively but lacks single-cell specificity.

 

In this study, we employ single-cell nanopore sequencing of full-length mRNAs across 27 glioblastoma patients, yielding approximately 210,000 single-cell transcriptomes. Integrating these results with paired short-read scRNA-seq allowed precise cell-type identification and detection of cell-type-specific isoforms. This approach enabled the discovery of cell-type-specific novel splicing events, including new combinations of known splice sites and undiscovered donor and acceptor sites, revealing a remarkable level of transcriptomic diversity within glioblastoma. This extensive single-cell analysis provides unprecedented insights into the transcriptional landscape of glioblastoma, identifying novel markers of transcriptional differentiation and potential therapeutic targets at single-cell resolution, advancing our understanding of splicing-driven glioblastoma heterogeneity and tumor progression.

 

 

Tay, Wan Jing

National University Hospital (Singapore)

Clinical and Molecular Divergences in Diffuse Midline Glioma, H3 K27-Altered: A Comparative Study of Survival Outcomes in Adults and Children

 

Background:

Diffuse midline glioma, H3 K27-altered (DMG), affects both adults and children. Previous studies reported conflicting data regarding survival differences between adults and children with DMG. This study compared clinical characteristics, molecular profiles, and survival outcomes between adult and paediatric patients with DMG.

 

Methods:

A pooled analysis of 1,498 patients (521 adults, 977 children) diagnosed with DMG was conducted using published data. The individual patient time-to-event data for overall survival (OS), defined as the time from diagnosis to death from any cause, were reconstructed from published Kaplan-Meier curves. Descriptive statistics were used to summarize the characteristics and molecular profiles. Cox regression was performed to compare OS between adults and children. Subgroup analyses were performed to evaluate potential factors that could modify the effect on OS between the two groups.

 

Results:

Brainstem involvement was less frequent in adults than in children (26% vs. 79%). Adults had higher rates of PPM1D (37% vs. 13%), FGFR1 mutations (17% vs. 6%), and NF1 mutations (35% vs. 12%) and lower rates of ACVR1 mutations (5% vs. 24%) than children. Adults had longer OS (median 16.2 months vs. 11.4 months, Hazard Ratio (HR) 0.52 (95% CI 0.46 – 0.59), p < 0.0001) than children. Subgroup analyses showed that brainstem involvement was a potential modifier on the effect of OS between adults and children (Brainstem: HR 0.47 (95% CI 0.33 – 0.66) vs No brainstem: HR 0.91 (95% CI 0.67 – 1.25), interaction P = 0.006). ATRX, NF1, TP53, and PPM1D mutations were not effect modifiers.

 

Conclusion:

The involvement of the brainstem, mutations in PPM1D, FGFR1, NF1, and ACVR1 genes differed between adults and children with DMG. Overall, adults had improved OS compared to children, especially in those with brainstem involvement. These findings highlight the need to further investigate the underlying biological differences in DMG between age groups.

 

Varlet, Pascale

GHU Paris-Psychiatrie et Neurosciences, Sainte-Anne Hospital

Methylation-based RTK2A and RTK2B diffuse pediatric high-grade gliomas present distinct radiological and histomolecular features

 

Diffuse pediatric-type high-grade gliomas (pedHGG), H3- and IDH-wildtype, encompass three main DNA-methylation-based subtypes: pedHGG-MYCN, pedHGG-RTK1A/B/C, and pedHGG-RTK2A/B. Since their first description in 2017 tumors of pedHGG-RTK2A/B have not been comprehensively characterized and their clinical significance is unknown. In a recent series of pedHGG with a gliomatosis cerebri (GC) growth pattern, an increased incidence of pedHGG-RTK2A/B (n=18) was observed. We added 14 epigenetically defined pedHGG-RTK2A/B tumors to this GC series and provided centrally reviewed radiological, histological, and molecular characterization. The final cohort of 32 pedHGG-RTK2A/B tumors consisted of 25 pedHGG-RTK2A (78%) and seven pedHGG-RTK2B (22%). The median age was 11.6 years (range, 4–17) with a median overall survival of 16.0 months (range 10.9–28.2). Seven of 11 added cases with imaging available showed a GC phenotype at diagnosis or follow-up. PedHGG-RTK2B tumors exhibited frequent bithalamic involvement (6/7, 86%). Central neuropathology review confirmed a diffuse glial neoplasm in all tumors with prominent angiocentric features in both subclasses. Most tumors (24/27 with available data, 89%) diffusely expressed EGFR with focal angiocentric enhancement. PedHGG-RTK2A tumors lacked OLIG2 expression, whereas 43% (3/7) of pedHGG-RTK2B expressed this glial transcription factor. ATRX loss occurred in 3/6 pedHGG-RTK2B samples with available data (50%). DNA sequencing (pedHGG-RTK2A: n=18, pedHGG-RTK2B: n=5) found EGFR alterations (15/23, 65%; predominantly point mutations) in both subclasses. Mutations in BCOR (14/18, 78%), SETD2 (7/18, 39%), and hTERT promoter (7/18, 39%) occurred exclusively in pedHGG-RTK2A tumors, while pedHGG-RTK2B tumors were enriched for TP53 alterations (4/5, 80%). In conclusion, pedHGG-RTK2A/B tumors revealed a highly diffuse-infiltrating tumor with specific radiological and histomolecular features. By comprehensive characterizing specific methylation-based tumors, the chance to facilitate specific and effective therapy concepts for these so far detrimental tumors increases.

 

 

Vella, Viviana

University of Sussex

Kinome-Wide Synthetic Lethal Screen Identifies PANK4 as a Modulator of Temozolomide Resistance in Glioblastoma

 

Background and Aims:

Temozolomide (TMZ) represents the cornerstone of glioblastoma (GBM) therapy. However, acquisition of resistance limits its therapeutic potential. The human kinome is an undisputable source of druggable targets, still, current knowledge remains confined to a limited fraction of it, with a multitude of under-investigated proteins yet to be characterised. In this study, we aimed to uncover synthetic lethal partners of TMZ in GBM that could enhance the effect of TMZ, thus improving TMZ therapy response.

 

Methods:

A kinome-wide RNAi screen was performed in a TMZ-resistant GBM cell model. A panel of several TMZ-resistant and patient-derived GBM cell lines was implemented for in vitro validation of our findings through several phenotypic assays. In vivo TMZ-resistant GBM xenografts and immunohistochemical (IHC) analysis of IDH-wildtype GBM specimens were employed to assess the clinical relevance of our findings. Tandem Mass Tag (TMT)-based quantitative proteomic studies were undertaken to elucidate the underlying mechanisms.

 

Results:

Following a kinome-wide RNAi screen, we identified pantothenate kinase 4 (PANK4) as a modulator of TMZ resistance in GBM and demonstrated that its depletion enhances the effect of TMZ, therefore improving the response to TMZ therapy. Validation of PANK4 across various TMZ-resistant GBM cell models and patient-derived GBM cell lines, immunohistochemical (IHC) analysis of IDH-wildtype GBM specimens, as well as in vivo studies, corroborated the potential translational significance of these findings. We also provided evidence that PANK4 expression is induced in response to TMZ treatment and increased PANK4 levels are associated with a worse clinical outcome. Moreover, by employing a Tandem Mass Tag (TMT)-based quantitative proteomic approach we revealed that PANK4 abrogation leads to a significant downregulation of numerous proteins with central roles in cellular detoxification and cellular response to oxidative stress. Furthermore, we showed that the ability of PANK4 to modulate sensitivity to TMZ treatment and intracellular reactive oxygen species (ROS) accumulation is dependent on its phosphatase activity. Mechanistically, as cells undergo genotoxic stress during TMZ exposure, PANK4 depletion represents a crucial event that can lead to accumulation of intracellular ROS and subsequent cell death.

 

Conclusion:

Collectively, our study provides novel insights into chemoresistance in GBM and unveils a previously unreported role for PANK4 in mediating therapeutic resistance to TMZ in GBM.

 

 

Wadekar, Ashish

All India Institute of Medical Sciences, New Delhi

Evaluating the efficacy of Hip1R, Vimentin, and H3K27Me3 as surrogate markers for 1p/19q codeletion in oligodendrogliomas

Introduction:

Assessment of the chromosomal 1p/19q status is essential to distinguish between IDH mutant astrocytoma and oligodendroglioma. Genetic analyses, however, are expensive, time-consuming, and not widely accessible. Immunohistochemical loss of ATRX is currently the only established surrogate marker for a non-1p/19q-codeleted genotype. To find cost-effective approaches and improve risk stratification, we aimed to investigate markers such as Vimentin, H3K27me3, and HIP1R.

Objective:

We aimed to assess the immunohistochemical expression HIP1R, Vimentin, and H3K27me3 as surrogate markers in predicting 1p/19q status in oligodendrogliomas.

Methods:

A total of 182 adult-type diffuse glioma patients were analyzed for IDH 1 R132H, ATRX, P 53, MIB1-LI, HIP1R, Vimentin, and H3K27me3 expression using immunohistochemistry. 1p/19q co-deletion was assessed by FISH assay. IDH sequencing was performed in IDH 1 R132H negative cases. Histomorphological and molecular classification of these gliomas was performed according to WHO 2021 CNS5 classification.

Results:

In this study 102 IDH-mutant oligodendrogliomas, 44 IDH-mutant astrocytomas, and 36 IDH-wild-type glioblastomas, exhibited distinct patterns of the IHC markers. In oligodendrogliomas, HIP1R showed either homogeneous or homogenous and mosaic staining, Vimentin was negative and H3K27me3 was lost in all cases. In IDH mutant astrocytomas and glioblastomas, HIP1R was predominantly mosaic, Vimentin was widely positive, and H3K27me3 was variable. Combining these markers, especially positivity of HIP1R, negative Vimentin, and complete loss of H3K27me3, achieved perfect diagnostic accuracy, making them highly reliable for differentiating oligodendrogliomas from astrocytomas, and glioblastomas.

Conclutions:

The study demonstrates that the combination of the three immunohistochemical markers HIP1R, Vimentin, and H3K27me3 can accurately predict 1p/19q co-deletion status in IDH-mutant gliomas. This method offers a reliable, robust and cost-effective alternative to complex techniques like FISH.

 

 

Wang, Da-Peng

Ruijin Hospital, Shanghai JiaoTong University

The m6A reader HNRNPC mediates BRD4 stabilization enhancing cabergoline efficacy in prolactinomas

 

Objective:

Pituitary adenomas (PAs), common intracranial benign tumors, include hormone-secreting functioning tumors and non-functioning pituitary adenomas. Prolactinomas (PRLomas) are usually treated with dopamine agonists (DAs), including cabergoline (CAB). Unfortunately, drug resistance occurs in approximately 20% of patients. Bromodomain protein 4 (BRD4), a transcriptional regulator that can be methylated by N6-methyladenosine (m6A), is involved in tumor growth and drug resistance, but its role in PRLomas is still unclear.

 

Methods:

Herein, the expression of BRD4 mRNA and protein was examined in PRLoma samples. The effect of silencing RNA against BRD4 (SiRNA-BRD4), treatment with CAB and combined treatment with both CAB and SiRNA-BRD4 on cell growth, apoptosis and PRL secretion were tested in MMQ and CH3 cells. Post-treatment mitochondrial oxidative stress and untargeted metabolomics were also evaluated. HNRNPC, BRD4, Scg2, DRD2 and apoptosis proteins were assessed by western blot. BRD4 methylation was investigated using m6A IP (MeRIP), dual-luciferase reporter and Actinomycin D assays. JQ-1, a BRD4 inhibitor, and CAB were used in eutopic and ectopic PRLoma animals.

 

Results:

BRD4 was highly expressed in PRLomas especially in patients with DA resistance, and it is positively related to drug resistance related genes, including TP53, SSTR2, PXN and KBTBD6. Inhibition of BRD4 promoted mitochondrial oxidative stress and dysbolism, apoptosis and PRL decline in cells. Compared to monotherapy, polytherapy had superior effects on anti-tumor growth and PRL suppression. SiRNA-BRD4 inhibited Scg2 expression but activated the DRD2 protein. Co-IP experiments confirmed the interactions between Scg2 and BRD4, Scg2 and DRD2. Meanwhile, the m6A reader HNRNPC methylated the 3’-untranslated region (UTR) but not the coding sequence (CDS) of BRD4, regulating its transcription.

 

Conclusion:

Inhibition of BRD4 can enhance the anti-tumor effects of CAB with a synergistic effect against PRLomas, which may be a viable therapeutic approach for DA-resistant patients.

 

 

Wang, Da-Peng

Fudan University

NKX2-5 as a promising prognostic indicator linked to tumor immune microenvironment in gliomas

 

Background:

NK2 homeobox 5 (NKX2-5) is a homeobox-containing transcription factor that exhibits differential expression in various cancers. However, its prognostic significance, influence on tumor immune infiltration, and potential mechanisms in glioma development remain elusive.

 

Methods:

We first conducted an analysis using data from The Cancer Genome Atlas (TCGA) and corroborated our findings with data from the Chinese Glioma Genome Atlas (CGGA). Univariate and multivariate evaluation of NKX2-5 in glioma prognosis. The UALCAN and MethHC databases were used to analyze the correlation between NKX2-5 and methylation, as well as methylation prognosis. The ssGSEA algorithm was further applied to analyze the association of NKX2-5 with immune cell infiltration, immune checkpoints, and drug sensitivity. GO and KEGG enrichment analyses were conducted to explore the potential biological functions and signaling pathways of the top 50 co-expressed genes with NKX2-5 in the TCGA database. RT-qPCR and immunohistochemistry experiments were used to detect the mRNA and protein expression levels of NKX2-5 in clinical tissue samples. MTS, EDU, Transwell, and wound healing assays were performed to evaluate the effects of NKX2-5 knockdown on the proliferation, invasion, and migration of glioma cells. Hoechst 33342 staining and flow cytometry were used to assess the effect of NKX2-5 deficiency on TMZ sensitivity in glioma cells, and ELISA was employed to measure IL6 and TNF expression levels.

 

Results:

Our univariate and multivariate Cox analyses confirmed that increased NKX2-5 mRNA levels are an independent predictor of poor prognosis. Glioma progression and stage were negatively associated with NKX2-5 promoter methylation. Interestingly, different methylation sites on the NKX2-5 gene were associated with distinct prognostic outcomes of glioma patients. Furthermore, High expression of NKX2-5 in glioma is positively correlated with macrophage infiltration and negatively correlated with CD8+ T cell infiltration. In addition, NKX2-5 may influence the tumor immune microenvironment and subsequently impact immune checkpoint activity, immune checkpoint blockade (ICB) therapy, and chemotherapy responses. Moreover, co-expression genes associated with NKX2-5 primarily function as extracellular matrix structural constituents and enzyme inhibitors, the signaling pathway involved in cell cycle, lysosome, and phagosome processes. Clinically, we confirmed the NKX2-5 is highly expressed in glioma and knockdown of NKX2-5 significantly reduced tumor proliferation and aggressive behavior. Additionally, NKX2-5 knockdown enhanced the sensitivity of glioma cells to the chemotherapeutic drug-temozolomide.

 

Conclusion Our findings suggest for the first time that NKX2-5 is a promising prognostic indicator for gliomas and a critical player in tumor immune microenvironment. These findings highlight its potential as a novel therapeutic target for future immunotherapy interventions.

 

 

Wei, Yanfei

Henan Academy of Sciences

Stalled oligodendrocyte differentiation in IDHmutant gliomas

 

Roughly 50% of adult gliomas harbor isocitrate dehydrogenase (IDH) mutations. According to the 2021 WHO classification guideline, these gliomas are diagnosed as astrocytomas, harboring no 1p19q co-deletion, or oligodendrogliomas, harboring 1p19q co-deletion. Recent studies report that IDH-mutant gliomas share a common developmental hierarchy. However, the neural lineages and differentiation stages in IDH-mutant gliomas remain inadequately characterized. Using bulk transcriptomes and single-cell transcriptomes, we identified genes enriched in IDH-mutant gliomas with or without 1p19q co-deletion, we also assessed the expression pattern of stage-specific signatures and key regulators of oligodendrocyte lineage differentiation. We compared the expression of oligodendrocyte lineage stage-specific markers between quiescent and proliferating malignant single cells. The gene expression profiles were validated using RNAscope analysis and myelin staining and were further substantiated using data of DNA methylation and single-cell ATAC-seq. As a control, we assessed the expression pattern of astrocyte lineage markers. Genes concordantly enriched in both subtypes of IDH-mutant gliomas are upregulated in oligodendrocyte progenitor cells (OPC). Signatures of early stages of oligodendrocyte lineage and key regulators of OPC specification and maintenance are enriched in all IDH-mutant gliomas. In contrast, signature of myelin-forming oligodendrocytes, myelination regulators, and myelin components are significantly down-regulated or absent in IDH-mutant gliomas. Further, single-cell transcriptomes of IDH-mutant gliomas are similar to OPC and differentiation-committed oligodendrocyte progenitors, but not to myelinating oligodendrocyte. Most IDH-mutant glioma cells are quiescent; quiescent cells and proliferating cells resemble the same differentiation stage of oligodendrocyte lineage. Mirroring the gene expression profiles along the oligodendrocyte lineage, analyses of DNA methylation and single-cell ATAC-seq data demonstrate that genes of myelination regulators and myelin components are hypermethylated and show inaccessible chromatin status, whereas regulators of OPC specification and maintenance are hypomethylated and show open chromatin status. Markers of astrocyte precursors are not enriched in IDH-mutant gliomas. These findings show that despite differences in clinical manifestation and genomic alterations, all IDH- mutant gliomas resemble early stages of oligodendrocyte lineage and are stalled in oligodendrocyte differentiation due to blocked myelination program. These findings provide a framework to accommodate biological features and therapy development for IDH-mutant gliomas (Genome Medicine (2023) 15:24).

 

 

Wong, Cheuk Lun

The University of Hong Kong

Yes-associated protein (YAP) modulation in targeting temozolomide chemoresistance in glioblastoma

 

The Hippo signaling pathway is often dysregulated in cancer. The study aims to investigate whether the Hippo signaling pathway contributes to temozolomide chemoresistance in glioblastoma, and to validate whether the combinational treatment of temozolomide and YAP inhibitor is effective against glioblastoma. Temozolomide-sensitive and temozolomide-resistant U87 and U251 GBM human cell lines were previously established. Temozolomide-resistant cells were maintained in low-dose temozolomide. Here, we investigate whether Hippo signaling contributes to temozolomide chemoresistance. By modulating the Hippo signaling pathway through short-hairpin YAP gene knockdown, the outcome on GBM cancer phenotypes was studied by cell assays (in vitro) and mouse orthotopic xenograft (in vivo). Finally, the therapeutic effect of YAP inhibitor is studied in vitro and in vivo respectively. Our data showed that YAP is overexpressed in temozolomide-resistant glioblastoma cells. Nuclear translocation of YAP (which becomes its active state) is increased in temozolomide-resistant cells. In fact, YAP inhibition demonstrates reduced cell viability and higher susceptibility to temozolomide. Mice with tumor injection of the YAP knockdown cells have significantly reduced tumor size. Similarly, YAP inhibitor resensitizes temozolomide-resistant cells to temozolomide. Mechanistically, YAP knockdown reduces cell stemness, which in turn attenuates temozolomide chemoresistance. In conclusion, our data suggests dysregulated Hippo signaling pathway contributes to temozolomide chemoresistance, thus targeting the Hippo signaling pathway resensitizes GBM cells to temozolomide. This research provides pre-clinical evidence on using YAP inhibitor for combinational therapy with temozolomide for the treatment of glioblastoma.

 

 

Wong, Ka Ho

LKS Faculty of Medicine, The University of Hong Kong

Immunogenicity Rescue of Tumor Microenvironment through the Manipulation of Low-density Lipoprotein Receptor-related Protein 1 in Tumor-Associated Macrophages in Glioblastoma

 

Purpose:

Glioblastoma (GBM) is the most common malignant primary brain tumor. The tumor microenvironment (TME) of GBM possesses a myeloid-dominant nature with T-cell scarcity, rendering traditional lymphoid-targeting immunotherapy ineffective. Myeloid-modifying treatment targeting tumor-associated macrophages (TAMs), historically described as immunosuppressive, is emerging as a promising alternative. In this study, we discovered a novel subpopulation of Low-density lipoprotein receptor-related protein 1 (LRP1)-high TAMs that exhibit anti-tumoral characteristics and showcased the therapeutic potential of modifying the LRP1 eat-me pathway in glioblastoma.

 

Methods:

Single-cell RNA sequencing (scRNA-seq) and spatial transcriptomic analysis was performed on clinical samples of glioblastoma patients. CX3CR1+/GFP and myeloid-depleted (CD11b-DTR) xenografts were utilized to assess in vivo macrophage characteristics and monitor tumor growth. Specifically, TAMs were subjected to in vivo LRP1 knockdown/overexpression through macrophage-specific AAV. Primary bone marrow-derived macrophages (BMDM) were generated from mouse bone marrow stromal cells, subjected to lentiviral knockdown/overexpression of LRP1, and subsequently used to reconstitute the macrophage environment of CD11b-DTR mice after myeloid depletion using diphtheria toxin. Tumors were collected from the xenografts for downstream analysis, including scRNA-seq, flow cytometry, immunofluorescence and immunohistochemical imaging, and protein/mRNA expression analysis. For in vitro experiments, knockdown of LRP1 and co-culture experiments with glioblastoma cells were performed with BMDM, human monocytic cell lines (U937, THP1), mouse macrophage cell line (RAW 246.7) and primary peripheral blood macrophages (PbMC) to assess in vitro phagocytosis and migration abilities.

 

Results:

scRNA-seq of glioblastoma patient samples reveals that LRP1 is highly expressed in pro-inflammatory (anti-tumoral) TAMs compared to anti-inflammatory (pro-tumoral) TAMs. LRP1-high TAMs demonstrate highly enriched anti-tumoral signaling pathways including phagocytosis and migration. Moreover, higher LRP1 expression in the TAMs is correlated with better patient survival. Spatial transcriptomic analysis identified the LRP1-calreticulin (one of the ""eat-me"" signaling pathways) as the major mode of interaction between LRP1-high TAMs and GBM tumor cells. In vivo xenografts demonstrated that AAV-mediated modification of LRP1 on TAMs leads to a stepwise impediment of tumor growth and enhancement of survival from LRP1 knockdown to control to overexpression. scRNA-seq analysis of tumors CD11b-DTR myeloid-depleted mice with BMDM reconstitution shows a more immunosuppressive TME landscape with LRP1-knockdown BMDM, and a more immune-activating TME landscape with LRP1-overexpressed BMDM, compared to control. In vitro co-culture experiments show that phagocytic rate is significantly reduced after LRP1 knockdown in macrophages. In addition, we identified vitamin D receptor (VDR) possesses transcriptomic control over LRP1 promoter through chromatin immunoprecipitation analysis, with LRP1 expression showing calcitriol (active vitamin D) dose-dependent increase on both mRNA and protein levels. Oral calcitriol treatment effectively promotes macrophage infiltration and phagocytosis in xenograft models.

 

Conclusions:

We hereby uncovered a novel LRP1-high subpopulation of TAMs showcasing strong anti-tumoral potentials, which is in stark contrast to the traditional notion of TAMs being immunosuppressive. We demonstrate the pivotal role of LRP1 in governing TAMs' anti-tumoral abilities in GBM. We also illustrated the therapeutic potential of myeloid-modulating treatments in GBM through LRP1 pathway enhancement using calcitriol treatment.

 

 

Xi, Shaoyan

Sun Yat-sen University Cancer Center

Comprehensive Analysis of Supratentorial Ependymoma, ZFTA-Fusion Positive with non-RELA Partners

 

The definition of ependymoma has moved from purely histopathological features to integrated diagnosis, relying heavily on molecular information. The 2021 World Health Organization (WHO) classification of central nervous system (CNS) tumors defines two molecular subtypes of supratentorial ependymoma (ST-EPN): one with zinc finger translocation-associated protein (ZFTA) fusion and another with yes-associated protein 1 (YAP1) fusion. The former has been previously named “ST-EPN, RELA-fusion positive” because ZFTA-RELA fusion was detected in most cases. However, ZTFA is now considered more representative than RELA because it may be fused with other partners. Here, we present ten additional cases of ST-EPN, ZFTA-fusion positive with non-RELA partners such as mastermind-like protein 2 (MAML2) (n=2), nuclear receptor coactivator 2 (NCOA2) (n=4), contactin-5 (CNTN5) (n=1), terminal uridylyl transferase 1 (TUT1) (n=1), synaptotagmin 12 (SYT12) (n=1), and transmembrane O-methyltransferase (LRTOMT) (n=1). Various histological characteristics were observed, including classic EPN-like, sarcoma-like, metastatic cancer-like, embryonal tumor-like, and even chordoma-like. These histological patterns brought out many differential diagnoses without next generational sequencing (NGS). DNA methylation profiling clustered the cases with the reference “EPN_ZFTA”. Together with the existing literature, we summarized 30 cases of ST-EPN, ZFTA-fusion positive with non-RELA partners. This tumor predominantly affected children and young adults without sexual predilection. Compared to ST-EPN with typical ZTFA-RELA fusion, tumors carrying non-RELA fusion partners showed a significantly better prognosis. Our study further expanded the morphologic and molecular genetic spectrum of ST-EPN, ZFTA-Fusion positive with non-RELA partners.

 

 

Yang, Ming

Cancer Hospital China Academy of Medical Sciences, National Cancer Centre China

The Safety and Efficacy of a cocktail therapy of Zanubrutinib, rituximab and High Dose- Methotrexate in treating Patients with Newly Diagnosed Primary Central Nervous System Lymphoma

 

Primary central nervous system lymphoma (PCNSL) is an aggressive non-Hodgkin lymphoma presenting with disease limited to the brain, spinal cord, leptomeninges, cerebrospinal fluid (CSF) and eyes. High-dose methotrexate (HD-MTX) chemotherapy is the preferred standard induction treatment regimen. We retrospectively studied the safety and efficacy of a combined cocktail therapy of Zanubrutinib, rituximab and HD-MTX as the induction therapy for PCNSL. The study enrolled a total of 99 patients with PCNSL treated in the Cancer Hospital Chinese Academy of Medical Science between 1st January 2021 and 31st December 2023. Among them, 18 patients received the cocktail therapy with rituximab, methotrexate and Zanubrutinib as the induction therapy. The safety and efficacy data were collected, including treatment response with follow up data, side effects, and prognosis. It has shown a promising result of the combined cocktail therapy as an induction treatment for PCNSL, which showed a favorable curative efficiency and milder adverse reactions. Moreover, a genetic analysis was performed. We analyzed the genetic profile associated with poor response and poor prognosis to the therapy. A few genetic footprints were identified associated with the poor response to the cocktail therapy. Thus, we propose the combined cocktail therapy of Zanubrutinib, rituximab and HD-MTX as the induction therapy for PCNSL is potentially a safe and effective option in treatment. And genetic screen may help to select the potential poor responders.

 

 

Yang, Ming

Cancer Hospital China Academy of Medical Sciences, National Cancer Centre China

Spinal Cord High Grade Glioma: A case series and review of the literature

 

In this paper, we present 2 cases of spinal HGG, one with diffuse midline glioma (DMG) and another patient with high grade astrocytoma, looking specifically into their histology subtypes, progress and treatment outcomes, along with a literature review.

 

Case study 1, a 35-year-old female presented with one month of neck pain after carrying heavy items. On examination she had left hemiparesis. MRI cervical spine revealed a large solitary enhancing intramedullary lesion from C2 to C5. She underwent C2-C6 laminectomy and debulking of the lesion. Histology showed high grade diffuse glioma, IDH wild type, with loss of staining for H3K27me3 and diffusely positive staining for H3K27M. Post operatively, she underwent fractionated radiotherapy (RT) with concurrent temozolomide (TMZ). Lomustine and bevacizumab was initiated, for which she is currently still receiving.

 

Case Study 2

A 49-year-old male presented with bilateral lower limb numbness, proprioceptive loss and urinary incontinence. MRI spine showed a T10–12 intramedullary lesion and he underwent a T11 laminectomy and biopsy. Histology returned as high grade astrocytoma, IDH1R132H negative, H3K27M negative. He underwent concurrent TMZ and RT with bevacizumab for 3 months, followed by adjuvant TMZ and Bevacizumab (BEV) for 1 year. However he developed worsening lower limb numbness and weakness and was switched to Lomustine/Bevacizumab for a year. Then his chemotherapy was changed to carboplatin and etoposide.This patient was alive at the 4 year follow up, however he developed extensive leptomeningeal disease with intracranial lesions and is currently undergoing palliative RT.

 

Literature Review

Evidence for the treatment of spinal HGG remains scarce and limited to mainly case reports. Most therapies are extrapolated from intracranial HGG studies, the standard of care being maximal safe resection followed by concurrent RT and TMZ for 6 weeks, then adjuvant TMZ for 6 months.

 

In one of the largest case series on primary spinal HGG of 29 patients, Upadhyay et al found no impact of concurrent chemotherapy on survival, with Temozolomide having limited evidence of benefit. However Temozolomide and Bevacizumab in particular have been shown to improve survival in retrospective series in recurrent HGGs, with Bevacizumab thought to decrease peritumoral oedema and mass effect. Outside of TMZ, there are three drugs and one device which are FDA- approved for the treatment of recurrent HGGs, namely Lomustine, Carmustine, Bevacizumab and tumour treating fields. Conversely, adjuvant RT has been shown to improve survival in patient with WHO grade 2-4 infiltrative astrocytoma, with the rate of local failure in spinal HGG of only 55% compared to over 80% for intracranial glioblastomas in the Stupp trial Recent trials have shown that convection enhanced delivery, blood-brain barrier opening, and immunotherapy are tolerated well by patients with H3K27M-mutated diffuse midline glioma, paving the way for future trials with novel agents and targeted therapies.

 

Conclusion

Prognosis of spinal HGG patient remains poor despite current standard of care with maximal safe resection and adjuvant chemoradiation. A better understanding of the molecular drivers of spinal HGGs and more clinical trials are needed to develop more effective treatment options.

 

 

Yao, Shun

The First Affiliated Hospital, Sun Yat-sen University

Leveraging Generative AI for MRI-to-CT Translation: Advancements and Applications in Neurosurgical Planning

 

Objective:

Computed tomography (CT) and magnetic resonance imaging (MRI) provide complementary insights for neurosurgical planning. While both modalities are valuable, obtaining both scans increases costs and exposes patients to additional ionizing radiation following an MRI. Cross-modal generation of high-resolution CT images from MRI data offers a promising alternative. This study aimed to develop deep learning-based frameworks, including a modified conditional generative adversarial network (cGAN) and a novel mask-guided dual network based on GAN architecture (MGDGAN), to synthesize CT (sCT) images from MRI scans.

 

Methods:

Pre-operative paired CT and contrast-enhanced MRI scans were collected from patients with conditions including meningiomas, pituitary tumors, vestibular schwannomas, and cerebrovascular diseases. The CT and MRI images were processed for denoising, field correction, and co-registration. MRI data were then used as input for the generative-AI frameworks, which generated sCT images. The accuracy of these synthetic images was assessed through quantitative similarity metrics and by evaluating clinical features, including measurements of the sella, internal auditory canal (IAC), and aeration patterns in the mastoid, clinoid, and sphenoid regions.

 

Results:

A total of 92,981 paired CT/MRI images from 80 patients were used for training and testing, with an additional 10,068 paired images from 10 patients serving as an external validation set. The sCT images successfully reconstructed the bony skull base and convexity with high accuracy. The median relative error in sella and IAC measurements between synthetic and real CT (rCT) scans was 6%, though there was greater variability in IAC reconstructions. Aeration in the mastoid, clinoid, and sphenoid regions was generally well-represented, although finer air cell septations showed some heterogeneity. Performance varied by pathology, with the most significant challenges in cases of meningiomas with intratumoral calcifications or calvarial invasion. Additionally, experimental results indicated that MGDGAN achieved high accuracy in generating fine bone structures, brain tissues, and cerebral lesions, producing images visually closer to rCT scans. Quantitative evaluations showed that MGDGAN outperformed other state-of-the-art methods, including CycleGAN, Pix2Pix, ECNN, cGAN9, APS, and ResViT across multiple datasets.

 

Conclusions:

The proposed generative AI frameworks, cGAN and MGDGAN, for MRI-to-CT translation have demonstrated significant potential for various neurosurgical applications, particularly as an adjunct tool for pre-operative planning of skull base surgeries. Further refinement of GAN variants performance for specific anatomical regions may further enhance its clinical utility.

 

 

"Yao, Shun

The First Affiliated Hospital, Sun Yat-sen University

MRI Atlas of Meningioma Spatial Distribution across Molecularly Integrated Grades

 

Background:

It is recognized that the site of meningioma origins across the meninges correlates with distinct symptoms, tissue histopathology subtypes, and surgical risks. However, pinpointing the exact location of the tumor's origin can be challenging, particularly in large tumors or those that span different brain areas. We employed an unbiased lesion mapping method to identify where meningiomas originate and how this relates to their biological characteristics and grading.

 

Methods:

We analyzed MRI scans, WHO grades, and a novel grading system called Integrated Grade (IG), which is based on genetic information, from 881 adult patients with confirmed intracranial meningiomas at a tertiary academic medical center. We utilized a semi-automated method to contour the tumors on pre-operative MRI scans and subsequently created a frequency atlas of meningioma locations using voxel-based lesion mapping (VBLM). This atlas illustrated the frequency of tumor occurrence in different areas and was compared to the tumors' origins.

 

 

Results:

Among the 881 patients (average age 57, 68.8% female), 589 had WHO grade 1 tumors (66.8%), 265 had WHO grade 2 (30.1%), and 27 had WHO grade 3 (3.1%). After reclassification based on molecular criteria, 585 were IG-1 (66.3%), 160 were IG-2 (18.2%), and 136 were IG-3 (15.5%). Less aggressive tumors were predominantly found in the middle of skull base and its surroundings. More aggressive meningiomas were more frequently observed in the falcine/parasagittal area and the right sphenoid region. When examining the molecular Integrated Grade, the clustering of tumors in certain areas was even more pronounced than when considering the WHO grade. WHO grade 2 tumors were distributed across IG 1-3, and their locations also varied accordingly.

 

Conclusion:

By employing the unbiased VBLM technique, we demonstrated that meningiomas exhibit a preference for specific areas, with certain areas being associated with more aggressive tumors. Different tumor distribution patterns were observed when considering both histopathological type and molecular grade. The molecular grade was associated with more distinct clusters of meningiomas, supporting the hypothesis that there is an association between the phenotype and genotypes of meningiomas.

 

 

Zhang, Qp

The 6th Affiliated Hospital of Shenzhen University Medical School

Brain cancer stem cells exosomes derived NamiRNA-enhancer network of miR-151a-3p activates PDE4D to promote Progression of glioma

 

Purpose:

Glioblastoma (GBM) is the most aggressive form of brain cancer, with a median survival time of less than 15 months despite multimodal treatment approaches. Brain tumor stem cells (BTSCs) contribute significantly to tumor recurrence and resistance to therapy. The molecular mechanisms by which BTSCs-derived exosomes influence glioma invasion and migration, particularly under hypoxic conditions, remain elusive. How miRNAs in the hypoxic BTSCs (H-BTSCs)-derived exosomes promoting the progression of glioma is currently unclear. Thus, studying these in depth would be beneficial in understanding their roles and mechanisms in different diseases.

 

Methods:

We conducted a comprehensive study involving high-throughput sequencing to identify differentially expressed miRNAs in exosomes derived from hypoxic BTSCs (H-BTSCs). We then employed Chip, dual luciferase reporter assays et al found that a NamiRNA-enhancer network of miR-151a-3p have the direct interaction between the phosphodiesterase 4D (PDE4D) protein. The study included in vitro assays using glioma cell lines, patient-derived xenograft (PDX) models, and biochemical analyses to assess the functional role of miR-151a-3p and PDE4D in glioma progression.

 

Results:

Our findings revealed a significant increase in miR-151a-3p levels under hypoxic conditions, which directly interacted with the PDE4D enhancer to activate the focal adhesion signaling pathway. miR-151a-3p upregulated PDE4D expression in glioma cells by enhancing the PDE4D promoter activity, forming a positive feedback loop that amplified glioma development. In vivo studies confirmed that H-BTSCs-derived exosomes rich in miR-151a-3p significantly enhanced tumor growth, invasion, and migration in orthotopic xenograft models.

 

Conclusion:

This study identifies a novel NamiR-151a-3p/PDE4D/focal adhesion kinase signaling axis that plays a critical role in the progression of glioblastoma. Our findings not only reveal a key mechanism by which hypoxic BTSCs-derived exosomes promote glioma invasion and migration but also provide a potential therapeutic target for the treatment of GBM. The identification of miR-151a-3p and PDE4D as central players in this process offers a new avenue for diagnostic and therapeutic intervention in glioblastoma.

 

 

Zheng, Danfeng

Third Hospital, School of Basic Medical Sciences, Peking University Health Science Center

A preliminary study on the clinical and pathological factors related to postoperative recurrence in patients with meningioma

 

Background:

Meningiomas are the most common primary tumor of the central nervous system, which are usually benign tumors that can be cured by surgical resection, but some patients relapse even after total surgical resection. How to solve the recurrence of meningiomas has always been a difficult problem in clinical work. Therefore, this study focused on exploring the clinical and pathological characteristics associated with meningioma recurrence after surgery, and screened the candidate gene KCNMA1 through the database to predict meningioma recurrence, and initially explored its protein expression in meningiomas and the molecular mechanism of its down-regulation in recurrent meningiomas.


Methods:

The clinical and pathological baseline data of 175 meningioma patients were collected in this study. Univariate and multivariate binary logistic regression were used to analyze the factors that influence patient recurrence. The differentially expressed gene KCNMA1 was screen out related to meningioma recurrence, and then KCNMA1 immunohistochemical staining was performed in 82 meningioma samples to explore its protein expression level in meningioma. Bisulfite PCR based on next-generation sequencing was used to detect specific DNA point mutations and promoter methylation levels of the KCNMA1 gene in paraffin-embedded meningioma tissue specimens.

Results:

1) recurrence occurred in 39 of 175 patients with meningiomas (recurrence rate 22.3%), including 23 of 144 patients with WHO grade 1 meningiomas (recurrence rate 16.0%). In the multivariate regression analysis, mitotic count ≥ 3/10 HPF (equivalent to 1.6mm2), WHO high grade and maximum tumor diameter ≥ 5 cm were prognostic factors for meningioma recurrence (P < 0.05). Moreover, in patients with WHO grade 1 meningiomas, the risk of recurrence of tumor with mitotic count ≥ 3/10 HPF and maximum diameter ≥ 5 cm is higher than that of overall meningiomas. 2) The expression level of KCNMA1 protein was down-regulated in recurrent meningiomas, and was an independent prognostic factor for recurrence of meningiomas included in multivariate analysis(overall: OR, 0.678, 95%CI, 0.570-0.807, P < 0.001; WHO grade 1: OR, 0.668, 95%CI, 0.530-0.843, P=0.001), and the expression level of KCNMA1 was negatively correlated with mitotic count (P=0.021). 3) Targeted sequencing of DNA samples showed that point mutations at major KCNMA1 sites (p.N999S, p.N536H, p.D434G) were not detected in recurrent meningiomas. However, the methylation levels of CpG_133, CpG_711, CpG_1859 and CpG_2526 in KCNMA1 promoter in recurrent meningiomas were higher than those in non-recurrent meningiomas, and the difference was statistically significant (P < 0.05).

Conclusions:

Mitotic count ≥ 3/10 HPF, high WHO grade and maximum tumor diameter ≥ 5 cm are prognostic factors for recurrence of meningiomas, while down-regulation of KCNMA1 protein expression is an independent prognostic factor for recurrence of meningiomas, and its molecular mechanism may be related to the hypermethylation of KCNMA1 promoter.