bims-malgli Biomed News
on Biology of malignant gliomas
Issue of 2021‒09‒05
thirteen papers selected by
Oltea Sampetrean
Keio University


  1. Front Cell Dev Biol. 2021 ;9 618961
      Among the most biologically, thus clinically, aggressive primary brain tumors are found malignant gliomas. Despite recent advances in adjuvant therapies, which include targeted and immunotherapies, after surgery and radio/chemotherapy, the tumor is recurrent and always lethal. Malignant gliomas also contain a pool of initiating stem cells that are highly invasive and resistant to conventional treatment. Ion channels and transporters are markedly involved in cancer cell biology, including glioma cell biology. Transient receptor potential (TRP) ion channels are calcium-permeable channels implicated in Ca2+ changes in multiple cellular compartments by modulating the driving force for Ca2+ entry. Recent scientific reports have shown that these channels contribute to the increase in glioblastoma aggressiveness, with glioblastoma representing the ultimate level of glioma malignancy. The current review focuses on each type of TRP ion channel potentially involved in malignant glioma cell death, with the ultimate goal of identifying new therapeutic targets to clinically combat malignant gliomas. It thus appears that cannabidiol targeting the TRPV2 type could be such a potential target.
    Keywords:  TRP ion channels; cannabidiol; cell death; malignant glioma; treatment
    DOI:  https://doi.org/10.3389/fcell.2021.618961
  2. Neurooncol Adv. 2021 Jan-Dec;3(1):3(1): vdab100
      Glioblastoma (GBM) is an incurable brain tumor with a median survival of approximately 15 months despite an aggressive standard of care that includes surgery, chemotherapy, and ionizing radiation. Mouse models have advanced our understanding of GBM biology and the development of novel therapeutic strategies for GBM patients. However, model selection is crucial when testing developmental therapeutics, and each mouse model of GBM has unique advantages and disadvantages that can influence the validity and translatability of experimental results. To shed light on this process, we discuss the strengths and limitations of 3 types of mouse GBM models in this review: syngeneic models, genetically engineered mouse models, and xenograft models, including traditional xenograft cell lines and patient-derived xenograft models.
    Keywords:  GL261; U87; glioblastoma; murine models; tumor
    DOI:  https://doi.org/10.1093/noajnl/vdab100
  3. Clin Cancer Res. 2021 Sep 03. pii: clincanres.1392.2021. [Epub ahead of print]
      PURPOSE: Glioblastoma (GBM) is an incurable primary brain tumor that has not benefited from immunotherapy to-date. Greater than 90% of GBM expresses the tryptophan (Trp) metabolic enzyme, indoleamine 2,3-dioxygenase 1 (IDO). This observation supported the historical hypothesis that IDO suppresses the antitumor immune response solely through a mechanism that requires intratumoral Trp depletion. However, recent findings led us to investigate the alternative hypothesis that IDO suppresses the anti-GBM immune response independent of its association with Trp metabolism.EXPERIMENTAL DESIGN: IDO-deficient GBM cell lines reconstituted with IDO wild-type or IDO enzyme-null cDNA were created and validated in vitro and in vivo Microarray analysis was conducted to search for genes that IDO regulates, followed by the analysis of human GBM cell lines, patient GBM and plasma, and the TCGA database. Ex vivo cell co-culture assays, syngeneic and humanized mouse GBM models were used to test the alternative hypothesis.
    RESULTS: Non-enzymic tumor cell IDO activity decreased the survival of experimental animals and increased the expression of complement factor H (CFH) and its isoform, factor H like protein 1 (FHL-1) in human GBM. Tumor cell IDO increased CFH and FHL-1 expression independent of tryptophan metabolism. Increased intratumoral CFH and FHL-1 levels were associated with poorer survival among glioma patients. Similar to IDO effects, GBM cell FHL-1 expression increased intratumoral Tregs and MDSCs while it decreased overall survival in mice with GBM.
    CONCLUSIONS: Our study reveals a newly non-metabolic IDO-mediated enhancement of CFH expression and provides a new therapeutic target in patients with GBM.
    DOI:  https://doi.org/10.1158/1078-0432.CCR-21-1392
  4. Neuro Oncol. 2021 Sep 01. 23(9): 1457-1469
      BACKGROUND: The DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT) causes resistance of tumor cells to alkylating agents. It is a predictive biomarker in high-grade gliomas treated with temozolomide, however, there is no consensus on which test method, methylation sites, and cutoff values to use.METHODS: We performed a Cochrane Review to examine studies using different techniques to measure MGMT and predict survival in glioblastoma patients treated with temozolomide. Eligible longitudinal studies included (i) adults with glioblastoma treated with temozolomide with or without radiotherapy, or surgery; (ii) where MGMT status was determined in tumor tissue, and assessed by 1 or more technique; and (iii) where overall survival was an outcome parameter, with sufficient information to estimate hazard ratios (HRs). Two or more methods were compared in 32 independent cohorts with 3474 patients.
    RESULTS: Methylation-specific PCR (MSP) and pyrosequencing (PSQ) techniques were more prognostic than immunohistochemistry for MGMT protein, and PSQ is a slightly better predictor than MSP.
    CONCLUSIONS: We cannot draw strong conclusions about use of frozen tissue vs formalin-fixed paraffin-embedded in MSP and PSQ. Also, our meta-analysis does not provide strong evidence about the best CpG sites or threshold. MSP has been studied mainly for CpG sites 76-80 and 84-87 and PSQ at CpG sites ranging from 72 to 95. A cutoff threshold of 9% for CpG sites 74-78 performed better than higher thresholds of 28% or 29% in 2 of the 3 good-quality studies. About 190 studies were identified presenting HRs from survival analysis in patients in which MGMT methylation was measured by 1 technique only.
    Keywords:   MGMT promoter methylation; glioblastoma; meta-analysis; prognostic biomarker; temozolomide
    DOI:  https://doi.org/10.1093/neuonc/noab105
  5. Neuro Oncol. 2021 Sep 02. pii: noab203. [Epub ahead of print]
      Glioblastoma is the most common primary malignant brain tumor in adults and it continues to have a dismal prognosis. The development of targeted therapeutics has been particularly challenging, in part due to a limited number of oncogenic mutations and significant intra-tumoral heterogeneity. TERT promoter mutations were first discovered in melanoma and later found to be present in up to 80% of glioblastoma samples. They are also frequent clonal alterations in this tumor. TERT promoter mutations are one of the mechanisms for telomerase reactivation, providing cancers with cellular immortality. Telomerase is a reverse transcriptase ribonucleoprotein complex that maintains telomere length in cells with high proliferative ability. In this article we present genomic and pre-clinical data that supports telomerase as a potential "Achilles' heel" for glioblastoma. We also summarize prior experience with anti-telomerase agents and potential new approaches to tackle this target.
    Keywords:  Glioblastoma; genomic driver; targeted therapies; telomerase
    DOI:  https://doi.org/10.1093/neuonc/noab203
  6. Clin Cancer Res. 2021 Sep 02. pii: clincanres.0544.2021. [Epub ahead of print]
      PURPOSE: Pyruvate kinase M2 (PKM2) catalyzes the final step in glycolysis, a key process of cancer metabolism. PKM2 is preferentially expressed by glioblastoma (GBM) cells with minimal expression in healthy brain. We describe the development, validation, and translation of a novel positron emission tomography (PET) tracer to study PKM2 in GBM. We evaluated 1-((2-fluoro-6-[18F]fluorophenyl)sulfonyl)-4-((4-methoxyphenyl)sulfonyl)piperazine ([18F]DASA-23) in cell culture, mouse models of GBM, healthy human volunteers, and GBM patients.EXPERIMENTAL DESIGN: [18F]DASA-23 was synthesized with a molar activity of 100.47 {plus minus} 29.58 GBq/µmol and radiochemical purity >95%. We performed initial testing of [18F]DASA-23 in GBM cell culture and human GBM xenografts implanted orthotopically into mice. Next we produced [18F]DASA-23 under FDA oversight, and evaluated it in healthy volunteers, and a pilot cohort of glioma patients.
    RESULTS: In mouse imaging studies, [18F]DASA-23 clearly delineated the U87 GBM from surrounding healthy brain tissue and had a tumor-to-brain ratio (TBR) of 3.6 {plus minus} 0.5. In human volunteers, [18F]DASA-23 crossed the intact blood-brain barrier and was rapidly cleared. In GBM patients, [18F]DASA-23 successfully outlined tumors visible on contrast-enhanced magnetic resonance imaging (MRI). The uptake of [18F]DASA-23 was markedly elevated in GBMs compared to normal brain, and it identified a metabolic non-responder within 1-week of treatment initiation.
    CONCLUSIONS: We developed and translated [18F]DASA-23 as a new tracer that demonstrated the visualization of aberrantly expressed PKM2 for the first time in human subjects. These results warrant further clinical evaluation of [18F]DASA-23 to assess its utility for imaging therapy-induced normalization of aberrant cancer metabolism.
    DOI:  https://doi.org/10.1158/1078-0432.CCR-21-0544
  7. Nat Commun. 2021 Sep 01. 12(1): 5203
      Aurora kinase A (AURKA) has emerged as a drug target for glioblastoma (GBM). However, resistance to therapy remains a critical issue. By integration of transcriptome, chromatin immunoprecipitation sequencing (CHIP-seq), Assay for Transposase-Accessible Chromatin sequencing (ATAC-seq), proteomic and metabolite screening followed by carbon tracing and extracellular flux analyses we show that genetic and pharmacological AURKA inhibition elicits metabolic reprogramming mediated by inhibition of MYC targets and concomitant activation of Peroxisome Proliferator Activated Receptor Alpha (PPARA) signaling. While glycolysis is suppressed by AURKA inhibition, we note an increase in the oxygen consumption rate fueled by enhanced fatty acid oxidation (FAO), which was accompanied by an increase of Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α). Combining AURKA inhibitors with inhibitors of FAO extends overall survival in orthotopic GBM PDX models. Taken together, these data suggest that simultaneous targeting of oxidative metabolism and AURKAi might be a potential novel therapy against recalcitrant malignancies.
    DOI:  https://doi.org/10.1038/s41467-021-25501-x
  8. Mol Cancer Ther. 2021 Aug 31. pii: molcanther.0547.2020. [Epub ahead of print]
      Glioblastoma (GBM) is the most common primary brain cancer in adults where tumor cell heterogeneity and sex differences influence clinical outcomes. Here, we functionally characterize three male and three female patient-derived GBM cell lines, identify pro-tumorigenic BTICs, and create novel male and female preclinical models of GBM. Cell lines were evaluated on the following features: proliferation, stemness, migration, tumorigenesis, clinical characteristics, and sensitivity to radiation, TMZ, rhTRAIL, and rhBMP4. All cell lines were classified as GBM according to epigenetic subtyping, were heterogenous and functionally distinct from one another, and re-capitulated features of the original patient tumor. In establishing male and female preclinical models, it was found that two male derived GBM cell lines (QNS108 and QNS120) and one female derived GBM cell line (QNS315) grew at a faster rate in female mice brains. One male derived GBM cell line (QNS108) decreased survival in female mice in comparison to male mice. However, no survival differences were observed for mice injected with a female derived cell line (QNS315). In summary, a panel of 6 GBM patient-derived cell lines were functionally characterized, and it was shown that BTIC lines can be used to construct sex-specific models with differential phenotypes for additional studies.
    DOI:  https://doi.org/10.1158/1535-7163.MCT-20-0547
  9. Nanoscale. 2021 Sep 02.
      MicroRNA-21 (miR-21) is involved in the progression of glioblastoma through inhibition of pro-apoptotic genes. Antisense RNA against miR-21 (antagomir-21) has been developed as a potential therapeutic reagent for the treatment of glioblastoma. The receptor for advanced glycation end-products (RAGE) is also involved in the progression of glioblastoma through induction of angiogenic factors. Therefore, RAGE-antagonist peptide (RAP) is proposed to be an anti-tumor reagent. In this study, self-assembled nanoparticles were produced solely with therapeutic agents, antagomir-21 and RAP, with no additional carrier. The therapeutic effects of the nanoparticles by intranasal delivery were evaluated in intracranial glioblastoma animal models. First, physical characterizations such as size/zeta-potential study, scanning electron microscopy, and gel retardation assays showed that antagomir-21 and RAP formed stable nanoparticles without any additional reagents. The ratio between antagomir-21 and RAP was optimized by an in vitro cellular uptake study. The antagomir-21/RAP nanoparticles were administrated intranasally in the intracranial glioblastoma animal models to bypass the blood-brain-barrier. As a result, the nanoparticles reduced the miR-21 levels in tumors. Inhibition of miR-21 by the nanoparticles induced the expression of pro-apoptotic genes, such as PTEN and PDCD4, which enhanced tumor cell apoptosis. In addition, the expression of RAGE was suppressed by the nanoparticles, resulting in decreased levels of vascular endothelial growth factor in the tumor. The reduction of CD31-positive endothelial cells confirmed the anti-angiogenic effects of the nanoparticles. The results indicate that the intranasal delivery of the self-assembled nanoparticles of antagomir-21 and RAP is an efficient treatment of glioblastoma.
    DOI:  https://doi.org/10.1039/d1nr03455c
  10. Neuro Oncol. 2021 Sep 03. pii: noab208. [Epub ahead of print]
      BACKGROUND: Telomere maintenance is increasingly recognised as being fundamental to glioma oncogenesis with longer leucocyte telomere length (LTL) reported to increase risk of glioma. To gain further insight into the relationship between telomere genetics and risk of glioma we conducted several complementary analyses, using GWAS data on LTL (78,592 individuals) and glioma (12,488 cases and 18,169 controls).METHODS: We performed both classical and Summary Mendelian Randomization (SMR), coupled with heterogeneity in dependent instruments tests, at genome-wide significant LTL loci to examine if an association was mediated by the same causal variant in glioma. To prioritize genes underscoring glioma-LTL associations we analysed gene expression and DNA methylation data.
    RESULTS: Genetically increased LTL was significantly associated with increased glioma risk, IVW-RE ORSD 4.79 (95% CI: 2.11-10.85, P = 1.76 × 10 -4). SMR confirmed the previously reported LTL associations at 3q26.2 (TERC; PSMR = 1.33 × 10 -5), 5p15.33 (TERT; PSMR = 9.80 × 10 -27), 10q24.33 (STN1 alias OBFC1; PSMR = 4.31 × 10 -5) and 20q13.3 (STMN3/RTEL1; PSMR = 2.47 × 10 -4) glioma risk loci. Our analysis implicates variation at 1q42.12 (PSMR = 1.55 × 10 -2), 6p21.3 (PSMR = 9.76 × 10 -3), 6p22.2 (PSMR = 5.45 × 10 -3), 7q31.33 (PSMR = 6.52 × 10 -3) and 11q22.3 (PSMR = 8.89 × 10 -4) as risk factors for glioma risk. Whilst complicated by patterns of linkage disequilibrium, genetic variation involving PARP1, PRRC2A, CARMIL1, POT1 and ATM-NPAT1 was implicated in the aetiology of glioma.
    CONCLUSIONS: These observations extend the role of telomere-related genes in the development of glioma.
    Keywords:  Mendelian Randomisation; Telomere length; glioma; risk factors
    DOI:  https://doi.org/10.1093/neuonc/noab208
  11. Sci Rep. 2021 Aug 31. 11(1): 17444
      Glioblastoma multiforme (GBM) is the most frequent type of primary astrocytomas. We examined the association between single nucleotide polymorphisms (SNPs) in Aurora kinase A (AURKA), Aurora kinase B (AURKB), Aurora kinase C (AURKC) and Polo-like kinase 1 (PLK1) mitotic checkpoint genes and GBM risk by qPCR genotyping. In silico analysis was performed to evaluate effects of polymorphic biological sequences on protein binding motifs. Chi-square and Fisher statistics revealed a significant difference in genotypes frequencies between GBM patients and controls for AURKB rs2289590 variant (p = 0.038). Association with decreased GBM risk was demonstrated for AURKB rs2289590 AC genotype (OR = 0.54; 95% CI = 0.33-0.88; p = 0.015). Furthermore, AURKC rs11084490 CG genotype was associated with lower GBM risk (OR = 0.57; 95% CI = 0.34-0.95; p = 0.031). Bioinformatic analysis of rs2289590 polymorphic region identified additional binding site for the Yin-Yang 1 (YY1) transcription factor in the presence of C allele. Our results indicated that rs2289590 in AURKB and rs11084490 in AURKC were associated with a reduced GBM risk. The present study was performed on a less numerous but ethnically homogeneous population. Hence, future investigations in larger and multiethnic groups are needed to strengthen these results.
    DOI:  https://doi.org/10.1038/s41598-021-96935-y
  12. Cancer Discov. 2019 Nov;9(11): 1476
      Cancer cells in the brain can form synaptic connections with neighboring neuronal cells, according to a trio of recently published studies. When activated, these synapses promote tumor proliferation, survival, and invasiveness. Disrupting the communication channels between neurons and cancer cells could help blunt the growth of deadly gliomas and brain metastases.
    DOI:  https://doi.org/10.1158/2159-8290.CD-NB2019-117
  13. Am J Clin Pathol. 2021 Aug 31. pii: aqab110. [Epub ahead of print]
      
    DOI:  https://doi.org/10.1093/ajcp/aqab110