bims-malgli Biomed News
on Biology of malignant gliomas
Issue of 2024‒01‒28
fifteen papers selected by
Oltea Sampetrean, Keio University



  1. bioRxiv. 2024 Jan 11. pii: 2023.04.04.535599. [Epub ahead of print]
      Pediatric high-grade gliomas are highly invasive and essentially incurable. Glioma cells migrate between neurons and glia, along axon tracts, and through extracellular matrix surrounding blood vessels and underlying the pia. Mechanisms that allow adaptation to such complex environments are poorly understood. N-cadherin is highly expressed in pediatric gliomas and associated with shorter survival. We found that inter-cellular homotypic N-cadherin interactions differentially regulate glioma migration according to the microenvironment, stimulating migration on cultured neurons or astrocytes but inhibiting invasion into reconstituted or astrocyte-deposited extracellular matrix. N-cadherin localizes to filamentous connections between migrating leader cells but to epithelial-like junctions between followers. Leader cells have more surface and recycling N-cadherin, increased YAP1/TAZ signaling, and increased proliferation relative to followers. YAP1/TAZ signaling is dynamically regulated as leaders and followers change position, leading to altered N-cadherin levels and organization. Together, the results suggest that pediatric glioma cells adapt to different microenvironments by regulating N-cadherin dynamics and cell-cell contacts.Summary: Pediatric gliomas invade the brain by migrating between nerve cells or exploiting extracellular matrix along blood vessels. This research reveals cross-talk between YAP1/TAZ signaling and N-cadherin that regulates leader-follower cell phenotypes and migration efficiency in neural and extracellular matrix environments.
    DOI:  https://doi.org/10.1101/2023.04.04.535599
  2. Acta Neuropathol. 2024 Jan 20. 147(1): 21
      The longitudinal transition of phenotypes is pivotal in glioblastoma treatment resistance and DNA methylation emerged as an important tool for classifying glioblastoma phenotypes. We aimed to characterize DNA methylation subclass heterogeneity during progression and assess its clinical impact. Matched tissues from 47 glioblastoma patients were subjected to DNA methylation profiling, including CpG-site alterations, tissue and serum deconvolution, mass spectrometry, and immunoassay. Effects of clinical characteristics on temporal changes and outcomes were studied. Among 47 patients, 8 (17.0%) had non-matching classifications at recurrence. In the remaining 39 cases, 28.2% showed dominant DNA methylation subclass transitions, with 72.7% being a mesenchymal subclass. In general, glioblastomas with a subclass transition showed upregulated metabolic processes. Newly diagnosed glioblastomas with mesenchymal transition displayed increased stem cell-like states and decreased immune components at diagnosis and exhibited elevated immune signatures and cytokine levels in serum. In contrast, tissue of recurrent glioblastomas with mesenchymal transition showed increased immune components but decreased stem cell-like states. Survival analyses revealed comparable outcomes for patients with and without subclass transitions. This study demonstrates a temporal heterogeneity of DNA methylation subclasses in 28.2% of glioblastomas, not impacting patient survival. Changes in cell state composition associated with subclass transition may be crucial for recurrent glioblastoma targeted therapies.
    Keywords:  DNA methylation; Deconvolution; Glioma; Outcome; Subgroup; Temporal
    DOI:  https://doi.org/10.1007/s00401-023-02677-8
  3. Front Immunol. 2023 ;14 1324618
      Glioblastoma (GBM) is the most aggressive and common type of malignant brain tumor diagnosed in adults. Preclinical immunocompetent mouse tumor models generated using mouse tumor cells play a pivotal role in testing the therapeutic efficacy of emerging immune-based therapies for GBMs. However, the clinical translatability of such studies is limited as mouse tumor lines do not fully recapitulate GBMs seen in inpatient settings. In this study, we generated three distinct, imageable human-GBM (hGBM) models in humanized mice using patient-derived GBM cells that cover phenotypic and genetic GBM heterogeneity in primary (invasive and nodular) and recurrent tumors. We developed a pipeline to first enrich the tumor-initiating stem-like cells and then successfully established robust patient-derived GBM tumor engraftment and growth in bone marrow-liver-thymus (BLT) humanized mice. Multiplex immunofluorescence of GBM tumor sections revealed distinct phenotypic features of the patient GBM tumors, with myeloid cells dominating the immune landscape. Utilizing flow cytometry and correlative immunofluorescence, we profiled the immune microenvironment within the established human GBM tumors in the BLT mouse models and showed tumor infiltration of variable human immune cells, creating a unique immune landscape compared with lymphoid organs. These findings contribute substantially to our understanding of GBM biology within the context of the human immune system in humanized mice and lay the groundwork for further translational studies aimed at advancing therapeutic strategies for GBM.
    Keywords:  BLT humanized mice; GBM - multiforme; flow cytometry; hGBM; humanized mice; multiplex immunofluorescence staining
    DOI:  https://doi.org/10.3389/fimmu.2023.1324618
  4. bioRxiv. 2024 Jan 08. pii: 2024.01.05.574065. [Epub ahead of print]
      Despite extensive advances in cancer research, glioblastoma (GBM) still remains a very locally invasive and thus challenging tumor to treat, with a poor median survival. Tumor cells remodel their microenvironment and utilize extracellular matrix to promote invasion and therapeutic resistance. We aim here to determine how GBM cells exploit hyaluronan (HA) to maintain proliferation using ligand-receptor dependent and ligand-receptor independent signaling. We use tissue engineering approaches to recreate the three-dimensional tumor microenvironment in vitro, then analyze shifts in metabolism, hyaluronan secretion, HA molecular weight distribution, as well as hyaluronan synthetic enzymes (HAS) and hyaluronidases (HYAL) activity in an array of patient derived xenograft GBM cells. We reveal that endogenous HA plays a role in mitochondrial respiration and cell proliferation in a tumor subtype dependent manner. We propose a tumor specific combination treatment of HYAL and HAS inhibitors to disrupt the HA stabilizing role in GBM cells. Taken together, these data shed light on the dual metabolic and ligand - dependent signaling roles of hyaluronan in glioblastoma.Significance: The control of aberrant hyaluronan metabolism in the tumor microenvironment can improve the efficacy of current treatments. Bioengineered preclinical models demonstrate potential to predict, stratify and accelerate the development of cancer treatments.
    DOI:  https://doi.org/10.1101/2024.01.05.574065
  5. Nat Commun. 2024 Jan 25. 15(1): 730
      Stimulating the innate immune system has been explored as a therapeutic option for the treatment of gliomas. Inactivating mutations in ATRX, defining molecular alterations in IDH-mutant astrocytomas, have been implicated in dysfunctional immune signaling. However, little is known about the interplay between ATRX loss and IDH mutation on innate immunity. To explore this, we generated ATRX-deficient glioma models in the presence and absence of the IDH1R132H mutation. ATRX-deficient glioma cells are sensitive to dsRNA-based innate immune agonism and exhibit impaired lethality and increased T-cell infiltration in vivo. However, the presence of IDH1R132H dampens baseline expression of key innate immune genes and cytokines in a manner restored by genetic and pharmacological IDH1R132H inhibition. IDH1R132H co-expression does not interfere with the ATRX deficiency-mediated sensitivity to dsRNA. Thus, ATRX loss primes cells for recognition of dsRNA, while IDH1R132H reversibly masks this priming. This work reveals innate immunity as a therapeutic vulnerability of astrocytomas.
    DOI:  https://doi.org/10.1038/s41467-024-44932-w
  6. Sci Rep. 2024 01 24. 14(1): 2123
      Quiescence, a reversible state of cell-cycle arrest, is an important state during both normal development and cancer progression. For example, in glioblastoma (GBM) quiescent glioblastoma stem cells (GSCs) play an important role in re-establishing the tumour, leading to relapse. While most studies have focused on identifying differentially expressed genes between proliferative and quiescent cells as potential drivers of this transition, recent studies have shown the importance of protein oscillations in controlling the exit from quiescence of neural stem cells. Here, we have undertaken a genome-wide bioinformatic inference approach to identify genes whose expression oscillates and which may be good candidates for controlling the transition to and from the quiescent cell state in GBM. Our analysis identified, among others, a list of important transcription regulators as potential oscillators, including the stemness gene SOX2, which we verified to oscillate in quiescent GSCs. These findings expand on the way we think about gene regulation and introduce new candidate genes as key regulators of quiescence.
    DOI:  https://doi.org/10.1038/s41598-024-51340-z
  7. Sci Rep. 2024 01 25. 14(1): 2126
      Identification of isocitrate dehydrogenase (IDH)-mutant glioma patients at high risk of early progression is critical for radiotherapy treatment planning. Currently tools to stratify risk of early progression are lacking. We sought to identify a combination of molecular markers that could be used to identify patients who may have a greater need for adjuvant radiation therapy machine learning technology. 507 WHO Grade 2 and 3 glioma cases from The Cancer Genome Atlas, and 1309 cases from AACR GENIE v13.0 datasets were studied for genetic disparities between IDH1-wildtype and IDH1-mutant cohorts, and between different age groups. Genetic features such as mutations and copy number variations (CNVs) correlated with IDH1 mutation status were selected as potential inputs to train artificial neural networks (ANNs) to predict IDH1 mutation status. Grade 2 and 3 glioma cases from the Memorial Sloan Kettering dataset (n = 404) and Grade 3 glioma cases with subtotal resection (STR) from Northwestern University (NU) (n = 21) were used to further evaluate the best performing ANN model as independent datasets. IDH1 mutation is associated with decreased CNVs of EGFR (21% vs. 3%), CDKN2A (20% vs. 6%), PTEN (14% vs. 1.7%), and increased percentage of mutations for TP53 (15% vs. 63%), and ATRX (10% vs. 54%), which were all statistically significant (p < 0.001). Age > 40 was unable to identify high-risk IDH1-mutant with early progression. A glioma early progression risk prediction (GlioPredictor) score generated from the best performing ANN model (6/6/6/6/2/1) with 6 inputs, including CNVs of EGFR, PTEN and CDKN2A, mutation status of TP53 and ATRX, patient's age can predict IDH1 mutation status with over 90% accuracy. The GlioPredictor score identified a subgroup of high-risk IDH1-mutant in TCGA and NU datasets with early disease progression (p = 0.0019, 0.0238, respectively). The GlioPredictor that integrates age at diagnosis, CNVs of EGFR, CDKN2A, PTEN and mutation status of TP53, and ATRX can identify a small cohort of IDH-mutant with high risk of early progression. The current version of GlioPredictor mainly incorporated clinically often tested genetic biomarkers. Considering complexity of clinical and genetic features that correlate with glioma progression, future derivatives of GlioPredictor incorporating more inputs can be a potential supplement for adjuvant radiotherapy patient selection of IDH-mutant glioma patients.
    DOI:  https://doi.org/10.1038/s41598-024-51765-6
  8. Clin Cancer Res. 2024 Jan 22.
      BACKGROUND: Adverse clinical events cause significant morbidity in patients with glioblastoma (GBM). We examined whether genomic alterations were associated with adverse events (AEs) in GBM patients.METHODS: We identified adults with histologically confirmed IDH-wildtype GBM with targeted next-generation sequencing (OncoPanel) at Dana Farber Cancer Institute from 2013-2019. Seizure at presentation, lymphopenia, thromboembolic events, pseudoprogression, and early progression (within 6 months of diagnosis) were identified as AEs. The biologic function of genetic variants was categorized as loss-of-function (LoF), no change in function, or gain-of-function (GoF) using a somatic tumor mutation knowledge base (OncoKB) and consensus protein function predictions. Associations between functional genomic alterations and AEs were examined using univariate logistic regressions and multivariable regressions adjusted for additional clinical predictors.
    RESULTS: 470 GBM patients met study criteria. 105 genes both had sequencing data available for ≥90% of patients and were altered in ≥10% of the cohort. Following false-discovery rate (FDR) correction and multivariable adjustment, the TP53, RB1, IGF1R, and DIS3 LoF alterations were associated with lower odds of seizures, while EGFR, SMARCA4, GNA11, BRD4, and TCF3 GoF and SETD2 LoF alterations were associated with higher odds of seizures. For all other AEs of interest, no significant associations were found with genomic alterations following FDR correction.
    CONCLUSION: Genomic biomarkers based on functional variant analysis of a routine clinical panel may help identify adverse events in GBM, particularly seizures. Identifying these risk factors could improve the management of patients through better supportive care and consideration of prophylactic therapies.
    DOI:  https://doi.org/10.1158/1078-0432.CCR-23-3018
  9. Neuro Oncol. 2024 Jan 22. pii: noae005. [Epub ahead of print]
      Randomized controlled trials (RCT) have been the gold standard for evaluating medical treatments for many decades but they are often criticized for requiring large sample sizes. For newly-diagnosed glioblastoma (GBM), the clinical trial landscape has seen little progress since the establishment of the standard of care (known as the "Stupp" regimen). Given the urgent need for better therapies, it has been argued that data collected from patients treated with the standard regimen can provide high-quality external control data to supplement or replace concurrent control arm in future RCT. The goal of this Review is to provide an in-depth appraisal of the use of external control data in the context of RCT. We describe several clinical trial designs with particular attention to how external information is utilized, and address common fallacies that may lead to inappropriate adoptions of external control data. Using two completed GBM trials, we illustrate the use of an assessment tool that lays out a blueprint for assembling a high-quality external control dataset. Using statistical simulations, we draw caution from scenarios where these approaches can fall short on controlling the type I error rate. Practical and statistical challenges associated with implementing these designs are also discussed.
    Keywords:  biases; concurrent control; external control; glioblastoma; type I error
    DOI:  https://doi.org/10.1093/neuonc/noae005
  10. Neuro Oncol. 2024 Jan 22. pii: noae012. [Epub ahead of print]
      BACKGROUND: This study evaluated whether generative artificial intelligence-based augmentation (GAA) can provide diverse and realistic imaging phenotypes and improve deep learning-based classification of isocitrate dehydrogenase (IDH) type in glioma compared with neuroradiologists.METHODS: For model development, 565 patients (346 IDH-wildtype, 219 IDH-mutant) with paired contrast-enhanced T1 and FLAIR MRI scans were collected from tertiary hospital and The Cancer Imaging Archive. Performance was tested on internal (119, 78 IDH-wildtype, 41 IDH-mutant [IDH1 and 2]) and external test sets (108, 72 IDH-wildtype, 36 IDH-mutant). GAA was developed using score-based diffusion model and ResNet50 classifier. The optimal GAA was selected in comparison with null model. Two neuroradiologists (R1, R2) assessed realism, diversity of imaging phenotypes, and predicted IDH mutation. The performance of a classifier trained with optimal GAA was compared with that of neuroradiologists using area under the receiver operating characteristics curve (AUC). The effect of tumor size and contrast enhancement on GAA performance was tested.
    RESULTS: Generated images demonstrated realism (Turing's test: 47.5%-50.5%) and diversity indicating IDH type. Optimal GAA was achieved with augmentation with 110 000 generated slices (AUC: 0.938). The classifier trained with optimal GAA demonstrated significantly higher AUC values than neuroradiologists in both the internal (R1, P=.003; R2, P<.001) and external test sets (R1, P<.01; R2, P<.001). GAA with large-sized tumors or predominant enhancement showed comparable performance to optimal GAA (internal test: AUC 0.956 and 0.922; external test: 0.810 and 0.749).
    CONCLUSIONS: Application of generative AI with realistic and diverse images provided better diagnostic performance than neuroradiologists for predicting IDH type in glioma.
    Keywords:  Artificial Intelligence; deep learning; glioma; isocitrate dehydrogenase; magnetic resonance imaging
    DOI:  https://doi.org/10.1093/neuonc/noae012
  11. Sci Rep. 2024 01 22. 14(1): 778
      Accurate determination of human tumor malignancy is important for choosing efficient and safe therapies. Bioimaging technologies based on luminescent molecules are widely used to localize and distinguish active tumor cells. Here, we report a human cancer grade probing system (GPS) using a water-soluble and structure-changeable Eu(III) complex for the continuous detection of early human brain tumors of different malignancy grades. Time-dependent emission spectra of the Eu(III) complexes in various types of tumor cells were recorded. The radiative rate constants (kr), which depend on the geometry of the Eu(III) complex, were calculated from the emission spectra. The tendency of the kr values to vary depended on the tumor cells at different malignancy grades. Between T = 0 and T = 3 h of invasion, the kr values exhibited an increase of 4% in NHA/TS (benign grade II gliomas), 7% in NHA/TSR (malignant grade III gliomas), and 27% in NHA/TSRA (malignant grade IV gliomas). Tumor cells with high-grade malignancy exhibited a rapid upward trend in kr values. The cancer GPS employs Eu(III) emissions to provide a new diagnostic method for determining human brain tumor malignancy.
    DOI:  https://doi.org/10.1038/s41598-023-50138-9