Nada Jabado's research while affiliated with SickKids and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (555)


LGG-13. UNRAVELING THE SPATIAL TUMOR MICROENVIRONMENT OF PEDIATRIC LOW-GRADE GLIOMAS USING IMAGING MASS CYTOMETRY
  • Article

June 2024

·

3 Reads

Neuro-Oncology

·

Romain Sigaud

·

Evan Puligandla

·

[...]

·

Pediatric low-grade gliomas (pLGG) are the most common brain tumors in children and are associated with significant morbidity. Therefore, there is an urgent need for novel therapeutic strategies. While some studies have described pLGG’s tumor microenvironment (TME) from bulk RNAseq and scRNAseq, little is known about the spatial architecture of pLGG and its association with clinico-molecular features. Our study utilized imaging mass cytometry and a panel of 35 metal-labelled antibodies to unravel the spatial organization of key TME cell populations in 120 primary pLGG samples from the LOGGIC Core BioClinical DataBank. Cellular neighborhood analysis was performed to map the spatial organization of pLGG TME. Several clinic-molecular features (entity, tumor location, genetic driver alteration, disease progression status, age and sex) were used to measure their putative association with the enrichment of key cell populations and cellular structures to identify diagnosis and prognosis markers. Here, we identified a predominant presence of myeloid cells in the TME, particularly notable in optic pathway tumors, which exhibited unique immune profiles. Optic pathway tumors demonstrated elevated immune subsets, delineating distinct architectural features in the TME of this brain region. Spatial analysis defined cellular neighborhoods and specific interactions thereof, including myeloid interaction and macrophage-abundant regions. Clinically, these myeloid cell populations, associated with an increased expression of the immune checkpoint protein TIM3, suggesting the presence of an immuno-suppressive environment, were associated with inferior survival outcomes. Importantly, we identified an immunophenotype signature based on the presence of 4 myeloid cell populations significantly associated with progression free survival in our cohort. Our study underscored the need for accurate identification of immune cell populations influencing tumor progression, offering valuable insights for the identification of prognostic markers, and for the development of effective therapeutic strategies, such as immune checkpoint inhibitors, for the treatment of pLGG.

Share

ETMR-28. EVALUATING THERAPEUTIC APPROACHES IN EMBRYONAL TUMORS WITH PLAGL1 AND PLAGL2 AMPLIFICATION AND UNRAVELING MOLECULAR PARALLELS IN EPIGENETICALLY RELATED TUMORS WITH PLAG1-ALTERATIONS

June 2024

·

3 Reads

Neuro-Oncology

BACKGROUND Embryonal tumors with PLAGL1 and PLAGL2 amplification (ET, PLAGL) display substantial clinical heterogeneity regarding applied treatment and outcomes. As a recently-defined entity, the spectrum of tumor-driving PLAG-family alterations is not yet fully elucidated. METHODS We analyzed clinical and MRI data from patients with ET, PLAGL. Second, we identified and analyzed tumors with epigenetic similarities. RESULTS 18 patients with ET, PLAGL revealed diverse clinical behavior. Patients with PLAGL1-amplified tumors (ET, PLAGL1) were older (median 8 years), had a higher rate of complete resections (6/9) and fewer relapses (3/9). Patients with PLAGL2-amplified tumors (ET, PLAGL2) were younger (median 1.9 years), often incompletely resected (6/9) and prone to earlier relapse/progression. Five-year PFS was 89% and 17% (ET, PLAGL1/ET, PLAGL2), with no predictive value of initial surgical extent on PFS/OS. Postoperative treatment included chemotherapy (17/18, various protocols) alone (n=8) or combined with RT (n=9). The three survivors with ET, PLAGL2 underwent induction and high-dose chemotherapy. All five patients with less intensive chemotherapy relapsed. Most first and all subsequent ET, PLAGL2 relapses were distant, questioning the value of initial local radiotherapy. Two ET, PLAGL1 relapses occurred >8 years after diagnosis (one after, one without previous RT). Through methylation-based t-SNE we identified 143 tumors with epigenetic similarities to ET, PLAGL. Filtering revealed a core set of 27 tumors, of which seven displayed evidence of PLAG1-fusion events and one PLAG1-amplification. RNAseq analysis (n=4) revealed distinct gene fusions involving PLAG1, with similar genes upregulated (RET, CYP2W1 and imprinted genes) as in ET, PLAGL. Like ET, PLAGL, PLAG1-fused tumors, occurred in young children with different diagnoses and localizations. CONCLUSION PLAG1-fused tumors are epigenetically similar to ET, PLAGL, show similarities in gene expression, and need to be differentiated from neuroepithelial tumor with PLAGL1-fusion. Future investigations will examine differences in clinical behavior and the utility of PLAG1/L1/L2 IHC for diagnosis.


LGG-40. TYPE II RAF INHIBITOR TOVORAFENIB IN RELAPSED/REFRACTORY PEDIATRIC LOW-GRADE GLIOMA (PLGG): REVERSIBLE DECREASES IN GROWTH VELOCITY IN THE PHASE 2 FIREFLY-1 TRIAL

June 2024

·

3 Reads

Neuro-Oncology

BACKGROUND: Tovorafenib is an investigational, selective, CNS-penetrant, type II RAF inhibitor. The ongoing FIREFLY-1 (NCT04775485) phase 2 study (Kilburn LK, et al. Nat Med. 2023) of tovorafenib in BRAF-altered pLGG resulted in antitumor activity and manageable safety. Decreased growth velocity (GV) was observed; this is an update on GV changes in skeletally immature children receiving tovorafenib. Methods A planned safety analysis was completed on August 8, 2023 on 137 patients (Arm 1: 77 & Arm 2: 60). Additional follow-up on all cases of decreased GV (an AESI) reported to the global safety database (GSDB) as of January 19, 2024 is provided. Results Overall, 29% had decreased GV from baseline (BL); 19% had ≥50% decrease. Of the 40 with this AESI, 75% had pre-existing neuromuscular or endocrine comorbidities potentially affecting normal growth, including 6 on GnRH-analogues for precocious puberty and 9 with BL heights 2 SDs above/below average for age and sex. Nineteen had on-treatment bone age assessments; none showed bone age advancement from BL or premature growth plate closure. No osteopenia or abnormal fractures reported. All 10 who discontinued or interrupted tovorafenib for ≥3 months for any reason (mean follow-up: 5.8 months), with off-treatment growth measurements available, showed post-treatment annualized GV (AGV) recovery (average AGV: on-treatment, 1.1 cm/y; off-treatment, 8 cm/y), with some exceeding expected average AGV for age. A 4-year-old boy with 1.2 cm/y AGV on-treatment had 12.3 cm/y AGV off-treatment (follow-up: 2 months). Five additional events of decreased GV in patients not on study FIREFLY-1 were reported to the GSDB; 4 of 5 had ≥3 months of off-treatment follow-up, all 4 recovered GV. Conclusions Decreased GV has been observed in patients on tovorafenib. Preliminary follow-up data in those who interrupted treatment show consistent evidence of GV recovery and preservation of growth potential on bone age studies.


HGG-39. CLINICAL CHARACTERISTICS AND SURVIVAL OUTCOMES OF DIFFUSE HEMISPHERIC GLIOMA, H3 G34-MUTANT: INTERIM RESULTS OF A MULTICENTRE INTERNATIONAL STUDY

June 2024

·

10 Reads

Neuro-Oncology

BACKGROUND Diffuse hemispheric glioma, H3 G34-mutant (G34-DHG) is generally associated with very poor outcome. Little is known, however, regarding long-term survivors and associated prognostic factors. METHODS This retrospective, multicentre study was designed to investigate clinical, molecular, and imaging variables that may impact the survival of patients with G34-DHG. Patients of any age diagnosed with G34-DHG after January 1, 1995, with at least one measure of survival or progression were eligible. RESULTS 69 patients with G34-DHG (62 G34R, 5 G34V, 2 diagnosed by methylation profiling only) have been included to date. Median age at diagnosis was 16.0 years (range: 2–38). 75% of patients were under the age of 18. 52% of patients were male. 8% of patients had multifocal/disseminated disease at diagnosis. Upfront therapy included gross/near total resection in 47% of patients, radiation therapy in 88% (89% focal, 6% CSI), and maintenance chemotherapy in 85% (62% TMZ-based, 33% TMZ/CCNU, 5% non-TMZ). Median follow-up time was 20.0 months (IQR=12.0–30.0). 86% of patients had progressive disease with a median time-to-progression of 14.0 months (IQR=6.6-22.5). At last follow-up, 70% of patients had died from disease, 23% were alive with disease, and 7% were alive without disease. Median overall survival was 24.7 months (IQR=18.8–30.6). Median time from progression to death was 6.0 months (IQR=2.0–10.8). Six patients were found to be long-term survivors > 5 years (four dead of disease, one alive with disease, one alive without disease). Log-rank assessment revealed improved survival to be associated with upfront gross/near-total resection (p=0.002) and age at diagnosis ≥ 18 years (p=0.012). CONCLUSIONS Surprisingly, we found that nearly 10% of G34-DHG patients survive 5 years or longer. Radical surgical resection and older age at diagnosis appear to be associated with longer survival. Imaging and molecular prognostic factors are being further investigated.


MDB-62. SUBGROUP AND SUBTYPE-SPECIFIC OUTCOMES IN METASTATIC INFANT MEDULLOBLASTOMA

June 2024

·

2 Reads

Neuro-Oncology

BACKGROUND Metastatic infant medulloblastoma (MB) is a major challenge given the devastating long-term effects of craniospinal irradiation to the developing brain. However large cohorts of metastatic infant medulloblastoma have not been analysed to date. METHODS A multicentre molecularly informed international cohort of children <6 years old with metastatic medulloblastoma was assembled and analysed. RESULTS Eighty-four patients with a median age of 2.95 (0.66-6.58) years were identified. 48.8% were Group3-MB, 23.8% Group4-MB and 27.4% SHH-MB. 78% of Group3-MB were subtype II (40.4%) and IV (38.1%), 50% of Group 4 were subtype VII, and within SHH, SHHß and SHHγ were 52.2% and 43.5% respectively. 28.5% of all Group3-MB harboured MYC amplification. A multivariable analysis incorporating subgroup, MYC amplification and upfront craniospinal irradiation revealed that survival was superior for SHH patients compared to Group 3 irrespective of MYC status (Group3-veMYCamp: HR 3.05 95%-CI 1.05-8.87 p=0.04, Group3+veMYCamp HR 7.87 95%-CI 2.49-24.87 p=0.00044). Group 4 had a trend to a poor outcome (HR 2.63 95%-CI 0.82-8.39 p=0.1). SHH subjects had a superior outcome with 5-year PFS of 0.657 (95%-CI 0.47-0.92), without a significant difference between SHHß and SHHγ. High-dose chemotherapy regimens portended to a superior outcome for both subtypes of SHH-MB. 5-year PFS for non-radiated SHH-MB was 61% (95%-CI 40.8-91.4), compared to non-radiated Group3-MB (18.5%; 95%-CI 5.4-62.8) and Group4-MB (20%; 95%-CI 3.4-100). SHH-MB patients had more local than metastatic relapses compared to non-SHH-MB (p=0.01). CONCLUSIONS In the largest study dedicated to metastatic infant medulloblastoma assembled to date, outcomes for infants with Group 3- and 4-MB with metastatic disease treated with radiation sparing approaches is dismal despite intensified regimens. MYC amplification portends to a near uniformly fatal prognosis with current treatment irrespective of upfront radiotherapy. Novel radiation sparing approaches are urgently needed for this group. Infants with both metastatic SHHß and SHHγ benefit from intensified therapy.


LGG-11. UNDERSTANDING THE TRANSCRIPTIONAL HETEROGENEITY OF PEDIATRIC LOW-GRADE GLIOMAS AND ITS IMPLICATION FOR TUMOR PATHOPHYSIOLOGY

June 2024

·

4 Reads

Neuro-Oncology

BACKGROUND Pediatric low-grade gliomas (pLGGs) are the most frequent brain tumors in children and comprise a heterogeneous group of tumors with different locations, histologic subtypes, ages at presentation, and clinical behavior. Tumors frequently respond to treatment with chemotherapy or radiation therapy, but they can regrow after a period of quiescence, requiring further therapy. Thus, a deeper understanding of the molecular processes involved in these tumors is required to develop therapeutic strategies that are effective against their disease mechanisms. METHODS To better understand the cellular behaviors of this heterogenous group of tumors, we have employed single-cell and single-nuclei RNA sequencing technologies to analyze a large-scale dataset (>250,000 cells) of 55 pLGGs across many histological subtypes (pilocytic astrocytoma, pleomorphic xanthoastrocytoma, pilomyxoid astrocytoma, DNET, ganglioglioma, RGNT, diffuse astrocytoma, SEGA, glioneuronal, oligodendroglioma). RESULTS Analysis of this data identified a heterogenous population of cell types and cell states, detecting mature and progenitor-like astrocytes and oligodendrocytes, as well as cells exhibiting senescence or cycling programs. Moreover, we identify a significant immune infiltrate, comprised primarily of microglia. In addition to heterogeneity within pLGG tumors, heterogeneity between LGG subtypes represents another layer that stratifies pLGG biology. We performed a compositional analysis of the cell types present in these tumors and compared transcription signatures and gene expression programs across shared cellular populations of histologically and genetically distinct pLGGs. Finally, we used spatial transcriptomic data to investigate and annotate the cellular architecture of multiple pLGGs with BRAF mutations and draw comparisons to findings from our single-cell and single-nuclei transcriptomic analysis. CONCLUSIONS Our analysis of human pLGGs at the single-cell and single-nuclei resolution provides critical insight into the heterogenous biological activities that constitute these tumors.


DIPG-76. PEDIATRIC PATIENTS WITH DIFFUSE MIDLINE GLIOMA DEMONSTRATE AN UNEXPECTED PREVALENCE OF GERMLINE VARIANTS IN HOMOLOGOUS RECOMBINATION GENES

June 2024

·

3 Reads

Neuro-Oncology

BACKGROUND Pediatric patients with Diffuse Midline Gliomas (DMG), H3K27M altered have a dismal prognosis and novel therapeutic approaches are urgently needed. Factors that drive development of pediatric DMG are unknown. METHODS To determine the prevalence of germline pathogenic/likely pathogenic variants (P/LPV) in DMG, we assembled an international cohort of 252 patients with germline whole genome or whole exome sequencing data, including diffuse intrinsic pontine glioma (DIPG; n=153), from Australian, European and North American centres. RESULTS We identified germline P/LPV in cancer predisposition genes in 7.5% (19/252) of patients, mainly homologous recombination (n=9; BRCA1, BRCA2, PALB2) and Fanconi anemia genes (n=4). Germline P/LPV in mismatch repair genes (MSH2, PMS2) were found in two patients. Two patients each had two separate germline P/LPV. The prevalence of germline P/LPV was not significantly different according to age, location of DMG nor H3K27M mutational status. Furthermore, tumor profiles differed, with absence of somatic drivers in the PI3K/mTOR pathway in patients with germline P/LPV compared to those without (P = 0.023). Knockdown of BRCA1 in DMG cell cultures sensitized tumor cells to PARP inhibition. Reflecting the potential therapeutic relevance of these findings, we describe one H3.3 K27M-mutant DMG patient with a pathogenic germline BRCA2 and FANCE variant and multiple recurrences, who was treated with a PARP inhibitor (olaparib) and immune checkpoint inhibitor, leading to a near complete radiological response after 4 months. CONCLUSION Our study is the largest series to date investigating germline P/LPV in cancer predisposition genes in DMG and provides new therapeutic insights. It is expected that these germline findings will also guide cascade testing for a proband’s relatives. Our data highlight the importance of germline testing in H3K27-altered DMG patients at diagnosis.


Epidemiology of Pediatric Tumors in Quebec: A 17-Year Report of Cancer in Young People in the Canada Registry
  • Article
  • Full-text available

May 2024

·

14 Reads

Current Oncology

Background. Cancer is the leading cause of disease-related death among children of more than 1 year of age. However, childhood cancer risk factors and etiology are yet to be fully understood. The goal of this study is to identify geographic variation among children and adolescents diagnosed with pediatric tumors between 2001 and 2018 in the province of Quebec. Methods. We analyzed pediatric patients less than 15 years of age from the Cancer in Young People in Canada (CYP-C) surveillance system who were diagnosed between 2001 and 2018 with cancer in the province of Quebec. The age-standardized age-adjusted incidence rates (AAIR) per 100,000 person years were calculated for all childhood cancers by cancer subgroups, Quebec Health regions, and age groups. Results. Overall, 3904 pediatric patients less than 15 years old were diagnosed with cancer in the province of Quebec in 2001–2018. The overall incidence rate (IR) in the province of Quebec was 16.14 (95%CL [15.56–16.73]) per 100,000 person years. For childhood cancers, regions that presented a higher AAIR were Chaudière-Appalaches and Capitale-Nationale with 18.2 and 17.5 per 100,000 person years, respectively. The incidence rates (IRs) in Chaudière-Appalaches (95% CI 1.0439–1.3532) and in Capitale-Nationale (95% CI 1.0124–1.2942) were statistically higher than the incidence in the province of Quebec (p = 0.0090 and p = 0.0310, respectively). When comparing the AAIR of the CNS tumor subgroup in Chaudière-Appalaches and in Capitale-Nationale, with the provincial average, we noticed a statistically higher incidence in Chaudière-Appalaches and a trend for Capitale-Nationale (p < 0.0001 and p = 0.0602, respectively). Conclusion. There is evidence of spatial clusters in Chaudière-Appalaches and Capitale-Nationale as areas for all childhood cancers. Further studies should be performed to investigate potential risk factors in these regions.

Download

The H3K27M mutation influences metabolic phenotypes and the metabolic response to radiation. A Untreated intracranial DIPGXIII xenografts and contralateral normal brain issue (n=2 mice/group) were harvested, and their metabolites were collected by methanol extraction and measured using LC/MS. Data are presented as the top 25 significantly different metabolites between Tumor and NB and are ordered by difference in average median centered abundance between H3K27M and H3K27M-KO cells. B Metabolite set enrichment analysis of all metabolites whose abundance were significantly different between Tumor and normal brain (83 total). Blue bars indicate pathways important for DMG-H3K27M biology as reported in the literature. Red bars indicate pathways important for purine metabolism. C Metabolite levels in untreated BT245 isogenic cell lines as measured using LC/MS. Data represent the top 25 significantly different metabolites between H3K27M-KO and H3K27M cells. Data are ordered by difference in median centered abundance in descending order. Metabolite extractions were performed in triplicate. D and E H3K27M-isogenic cell lines DIPGXIII (D) and BT245 (E) were treated with or without 4Gy RT and harvested for metabolite quantification 2hrs later. Metabolites meeting the FC threshold were selected. Data represent the top 25 largest absolute post-RT abundance differences between H3K27M and H3K27M-KO cells in descending order. Red indicates abundances over a post-RT FC value of 0.0. Blue indicates abundances under a post-RT FC value of 0.0. F and G Metabolic set enrichment analysis was performed on the metabolite lists for DIPGXIII (D) and BT245 (E) and ordered based on –log10p-value. Red bars indicate pathways important for purine metabolism. H Venn Diagram depicting the metabolic pathways enriched in RT-treated DMG-H3K27M isogenic cell lines. Bold-faced font indicates metabolic pathways important for purine metabolism and synthesis. I Purine metabolites from DIPGXIII xenograft tumors and contralateral normal brain from A) were compared and ordered by difference in average median centered abundance between tumor and normal brain
H3K27M cells have highly active purine synthesis. A Schematic of purine synthetic metabolic pathways showing where the purine de novo and salvage pathways converge to form mature purines. Schematic was created using BioRender.com. B-E Total abundance of ¹³C-labeled (B) F16BP, (C) R5P, (D) GMP, and (E) AMP in H3K27M cells when normalized to H3K27M-KO cells. F Total abundance of ¹⁵N-Gln-labeled GMP (left) and AMP (right) in H3K27M-isogenic cells. H Total abundance of deuterium-labeled GMP (left) and AMP (right) in H3K27M-isogenic cells. H Ratio of ¹⁵N-Gln:2D-Hpx labeled relative abundance of GMP (left) and AMP (right) in untreated DIPGXIII H3K27M-isogenic cell lines. Statistical analyses were performed using two-tailed t-tests in GraphPad Prism 10.0
De novo purine synthesis inhibition radiosensitizes K27M cells. A and B Immunoblots of (A) IMPDH1 and (B) HGPRT expression in DMG-H3K27M isogenic paired cell lines. Densitometric values were calculated using ImageJ software and expression values were normalized to H3K27M-KO cells. Expression ratios are listed below the blot images. C Publicly available RNAseq Z-score data for HPRT1 transcript expression from pediatric high-grade gliomas (pHGG) was obtained through PedCBioPortal and filtered to include only samples with known H3 mutational status (for both H3F3A and HIST1H3B) to include all known H3WT (n=59) and combined H3K27M (H3F3A-mut+HIST1H3B-mut) samples (n=44).D Schematic depicting the hypothesis that the K27M mutation induces defective guanylate purine salvage through HGPRT suppression, leaving K27M cells vulnerable to de novo guanylate synthesis using IMPDH inhibition using MPA. Schematic was created using BioRender.com. E And G Long-term neurosphere growth assays for (E) DIPGXIII and (G) BT245 H3K27M-expressing cell lines treated with increasing doses of RT (0, 2, 6Gy) with or without 1-10μM MPA (left) and corresponding enhancement ratio (Dbarcontrol/DbarTx) for each concentration of MPA (right) administered with RT. Each long-term neurosphere assay was performed 3x per cell line. Statistical analyses were performed using two-tailed t-tests in GraphPad Prism 10.0. F and H Live cell imaging analysis of (F) DIPGXIII and (H) BT245 neurospheres treated with 0-6Gy +/- 10μM MPA. Images taken 11 days (DIPGXIII) and 9 days (BT245) after replating in 96-well plates. Images acquired using a Cytation 5 plate reader and attached BioSpa incubator (Agilent Technologies)
Combination RT and de novo purine synthesis inhibition reduces DMG-H3K27M tumor size and extends mouse survival in vivo. A Schematic of treatment schedule administered to DIPGXIII-bearing Rag1-KO mice. DIPGXIII cells expressing GFP and Luciferase (LUC) (DIPGXIII-GFP/LUC) were orthotopically implanted into the cortex and mice were administered 150mg/kg MMF for 11 days, with 6 intermittent doses of 2Gy RT (red bolts). Schematic was created using BioRender.com. B Spider plot of bioluminescent signal flux values for individual DIPGXIII-GFP/LUC mouse tumors. Blue box indicates the treatment period. C Representative bioluminescent signal images for one mouse per treatment group 24 days after implantation. D Kaplan-Meier survival analysis of Rag1-KO mice bearing treated and untreated DIPGXIII-GFP/LUC orthotopic xenograft tumors (n=10 mice/group). Statistical analysis was performed by comparing individual survival curves of individual treatment groups in GraphPad Prism 10.0
H3K27M-expressing cells increase hypoxanthine-derived salvage and maintain high guanine salvage following RT. A and B Immunoblots of (A) IMPDH1 and (B) HGPRT expression before and after 4Gy RT in DIPGXIII cells. Densitometric analysis was performed using ImageJ software and expression values were normalized to No-RT controls. Values are listed below the blot images. C FC in the ion abundances of ¹⁵N-labeled GMP in DIPGXIII H3K27M-isogenic cell lines before (gray bars) and 3hrs after 4Gy RT (red bars). Data are normalized to untreated control samples. D FC in the ion abundances of 1D-labeled GMP in DIPGXIII H3K27M-isogenic cell lines before (gray bars) and 3hrs after 4Gy RT (red bars). Data are normalized to untreated control samples. E Ratio of ¹⁵N-Gln:2D-Hpx label incorporation into GMP after 3 hours in DIPGXIII cells that were either untreated or treated with 4Gy RT. Data is normalized to untreated control. F Total abundance of ¹³C8-labeled GMP in H3K27M-isogenic cells. G FC in the ion abundances of ¹³C8-labeled GMP in DIPGXIII H3K27M-isogenic cell lines before (gray bars) and 3hrs after 4Gy RT (red bars). H Percent relative abundances of 1D-labeled GMP (light blue bars) and ¹³C8-labeled GMP (red bars) in H3K27M-isogenic cells before and after 3hrs 4Gy RT. Statistical analyses were performed using two-tailed t-tests in GraphPad Prism 10.0

+1

Purine salvage promotes treatment resistance in H3K27M-mutant diffuse midline glioma

April 2024

·

48 Reads

Cancer & Metabolism

Background Diffuse midline gliomas (DMG), including diffuse intrinsic pontine gliomas (DIPGs), are a fatal form of brain cancer. These tumors often carry a driver mutation on histone H3 converting lysine 27 to methionine (H3K27M). DMG-H3K27M are characterized by altered metabolism and resistance to standard of care radiation (RT) but how the H3K27M mediates the metabolic response to radiation and consequent treatment resistance is uncertain. Methods We performed metabolomics on irradiated and untreated H3K27M isogenic DMG cell lines and observed an H3K27M-specific enrichment for purine synthesis pathways. We profiled the expression of purine synthesis enzymes in publicly available patient data and our models, quantified purine synthesis using stable isotope tracing, and characterized the in vitro and in vivo response to de novo and salvage purine synthesis inhibition in combination with RT. Results DMG-H3K27M cells activate purine metabolism in an H3K27M-specific fashion. In the absence of genotoxic treatment, H3K27M-expressing cells have higher relative activity of de novo synthesis and apparent lower activity of purine salvage demonstrated via stable isotope tracing of key metabolites in purine synthesis and by lower expression of hypoxanthine-guanine phosphoribosyltransferase (HGPRT), the rate-limiting enzyme of purine salvage into IMP and GMP. Inhibition of de novo guanylate synthesis radiosensitized DMG-H3K27M cells in vitro and in vivo. Irradiated H3K27M cells upregulated HGPRT expression and hypoxanthine-derived guanylate salvage but maintained high levels of guanine-derived salvage. Exogenous guanine supplementation decreased radiosensitization in cells treated with combination RT and de novo purine synthesis inhibition. Silencing HGPRT combined with RT markedly suppressed DMG-H3K27M tumor growth in vivo. Conclusions Our results indicate that DMG-H3K27M cells rely on highly active purine synthesis, both from the de novo and salvage synthesis pathways. However, highly active salvage of free purine bases into mature guanylates can bypass inhibition of the de novo synthetic pathway. We conclude that inhibiting purine salvage may be a promising strategy to overcome treatment resistance in DMG-H3K27M tumors.


Very Long-Term Survivorship in Pediatric DIPG: Case Report and Review of the Literature

April 2024

·

22 Reads

Journal of Pediatric Hematology/Oncology

Diffuse intrinsic pontine gliomas are lethal tumors with a prognosis generally less than 1 year. Few cases of survivors of 5 years or more have been reported. This case report highlights the journey of a 9.5-year survivor who underwent 3 rounds of focal radiotherapy; she experienced 6 years of progression-free survival following the first round but ultimately succumbed to her disease. An autopsy revealed a favorable IDH1 mutation and the absence of H3K27M. This case reiterates the importance of extensive molecular analyses in diffuse intrinsic pontine gliomas and explores the potential benefit of re-irradiation in patients with positive responses and long periods of remission.


Citations (42)


... Phenotypic variability could also be affected by which protein domain, specifically the tail or core, is affected by the variant. Certain "hotspot" somatic variants in H3.3, which are restricted to residues that include H3.3 p.K27M and p.G34R/V, cause pediatric brain tumors [17][18][19][20][21][22]. While these somatic variants are restricted to the H3.3 tail, the causative germline variants in BLBS are distributed throughout H3.3 (Fig. 1B). ...

Reference:

Expanded phenotypic spectrum of neurodevelopmental and neurodegenerative disorder Bryant-Li-Bhoj syndrome with 38 additional individuals
Pediatric glioma histone H3.3 K27M/G34R mutations drive abnormalities in PML nuclear bodies

Genome Biology

... In recent years, targeted therapies for pLGG have become available [7][8][9] which are beyond the focus of this paper. Certainly, efficacy reports on MAPK inhibitors open new perspectives on tumor volume reduction [20,21]. However, treatment-related adverse events were reported in a relevant portion of patients (42% ≥ grade 3), and 18.2% discontinued treatment due to adverse events or any kind of progression. ...

The type II RAF inhibitor tovorafenib in relapsed/refractory pediatric low-grade glioma: the phase 2 FIREFLY-1 trial

Nature Medicine

... It was suggested that further treatment regimens, enhanced drug delivery methods and combination therapies should be considered in future clinical studies using panobinostat. 129 Other clinical studies that use panobinostat include a phase 1 trial using Marizomib, a proteasome inhibitor in combination with panobinostat in pHGGs (NCT04341311) and phase 1/2 trials that are looking at the safety profile and drug tolerability of MTX110, a water soluble nanoformulation of panobinostat, in children newly diagnosed with DIPG (NCT03566199 and NCT04264143). Some other clinical trials using HDACi to treat pA that are currently underway/completed or in the recruiting phase are with vorinostat (class 1, 2 and 4 HDACi) alone or given together with RT (NCT01189266) or in combination with temsirolimus, a mTOR kinase inhibitor and RT (NCT02420613) and valproic acid (NCT00107458). ...

A Phase I Trial of Panobinostat in Children with Diffuse Intrinsic Pontine Glioma: A Report from the Pediatric Brain Tumor Consortium (PBTC-047)
  • Citing Article
  • August 2023

Neuro-Oncology

... Importantly, the high error rate observed in these devices can be attributed to the low sequencing depth of LRS, with reported sequencing error rates of 10-15% in SMRT and 5-20% in nanopore sequencing (199). One of the solutions for this drawback is the implementation of short-reading sequencing with long-reading sequencing, as it can importantly improve data analysis by having better accuracy, such as in the study did by Zwaig et al. (137). As most genetic studies on cancer have focused on other more used and wellknown technologies such as NGS, there is a necessity for the continuous improvement of LRS in terms of tools for data analysis such as the elaboration of new algorithms for better analysis of longer and complex reads. ...

Linked-read based analysis of the medulloblastoma genome
Frontiers in Oncology

Frontiers in Oncology

... Interestingly, sensitivity to MAPK inhibitor treatment seemed to be mainly influenced by immune infiltration, again emphasizing the importance of the neuro-immunologic crosstalk. 26,27 ...

MAPK inhibitor sensitivity scores predict sensitivity driven by the immune infiltration in pediatric low-grade gliomas

Nature Communications

... The total diagnostic interval was 8 (3,16) weeks with no statistically significant difference between the patients with germline variants (11.5 (2.75, 32.75) weeks) and patients without them (8 (3, 14) weeks). ...

Performance of the eHealth decision support tool, MIPOGG, for recognising children with Li-Fraumeni, DICER1, Constitutional mismatch repair deficiency and Gorlin syndromes
  • Citing Article
  • July 2023

Journal of Medical Genetics

... Recently, the combination of dabrafenib-trametinib resulted in significantly more responses and longer PFS in pLGG patients with BRAFV600 mutations when compared to standard chemotherapy (carboplatin plus vincristine) in the first-line non-surgical therapy setting [26]. Following on from this trial, there are at present at least four ongoing clinical trials for BRAF altered or other pLGG types with some of them excluding V600E BRAF mutation, that are randomizing between standard chemotherapy and targeted therapy as first-line non-surgical therapy for LGGs (LOGGIC--Firefly 2 [27], PLGG-METRIC in France, COG trials ACNS1831 for NF1-related LGGs and ACNS1833). ...

LOGGIC/FIREFLY-2: A phase 3, randomized trial of tovorafenib vs. chemotherapy in pediatric and young adult patients with newly diagnosed low-grade glioma harboring an activating RAF alteration.
  • Citing Article
  • June 2023

Journal of Clinical Oncology

... These models were able to evaluate the H3.3 G34R mutation with its associated mutation in the PDGFRA gene, showing that G34R DHGs can arise from the ganglionic eminences (similar to the germinal matrices of human embryos), as well as generating mouse models for H3K27M with mutations in NF1, FGFR1, PPM1D, PIK3CA, and ACVR1. In addition to targeting these cancers in a spatiotemporal context, models incorporating IUE methods allow for the evaluation of these cancers of development to be investigated in the developing brain, which contrasts to previous models in which tumors grow in adult mice [85]. ...

A Compendium of Syngeneic, Transplantable Pediatric High-Grade Glioma Models Reveals Subtype-Specific Therapeutic Vulnerabilities

Cancer Discovery

... Recognition of the stagnating reduction of childhood cancer mortality rates 8 , coupled with increasingly accessible and sophisticated next generation sequencing (NGS) and bioinformatics, has fueled a new age of molecular discovery in pediatric oncology. Access to comprehensive genomic analyses has rapidly accelerated with the establishment of multiple, large pediatric cancer precision medicine programs [9][10][11][12][13][14][15][16][17][18] . These trials demonstrate the utility of NGS assays in multiple categories of actionability including refining diagnoses, discovery of underlying cancer predisposition in 7-18% of participants and identifying therapeutically actionable variants in up to 70-86%, with variable level of evidence (LOE) schema. ...

Abstract 4509: A pan-Canadian precision oncology program for children, adolescents and young adults with hard-to-cure cancer: The PRecision Oncology For Young peopLE (PROFYLE) Program
  • Citing Article
  • April 2023

Cancer Research

... Currently reported DNMT3A variants associated with TBRS have not been found in our study. Recent research has revealed potential roles of DNMT3A and histones in in mediating progressive neurodegeneration through the regulation of synaptic pruning and neuroinflammation [32]. The TET family of methylcytosine dioxygenases comprises three members, TET1, TET2, and TET3, which catalyze the oxidation of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), initiating active DNA demethylation through the base-excision repair pathway [33]. ...

Single substitution in H3.3G34 alters DNMT3A recruitment to cause progressive neurodegeneration
  • Citing Article
  • March 2023

Cell