Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 194
Filter
1.
J Control Release ; 370: 835-865, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38744345

ABSTRACT

Diffuse midline glioma (DMG), including tumors diagnosed in the brainstem (diffuse intrinsic pontine glioma - DIPG), is the primary cause of brain tumor-related death in pediatric patients. DIPG is characterized by a median survival of <12 months from diagnosis, harboring the worst 5-year survival rate of any cancer. Corticosteroids and radiation are the mainstay of therapy; however, they only provide transient relief from the devastating neurological symptoms. Numerous therapies have been investigated for DIPG, but the majority have been unsuccessful in demonstrating a survival benefit beyond radiation alone. Although many barriers hinder brain drug delivery in DIPG, one of the most significant challenges is the blood-brain barrier (BBB). Therapeutic compounds must possess specific properties to enable efficient passage across the BBB. In brain cancer, the BBB is referred to as the blood-brain tumor barrier (BBTB), where tumors disrupt the structure and function of the BBB, which may provide opportunities for drug delivery. However, the biological characteristics of the brainstem's BBB/BBTB, both under normal physiological conditions and in response to DIPG, are poorly understood, which further complicates treatment. Better characterization of the changes that occur in the BBB/BBTB of DIPG patients is essential, as this informs future treatment strategies. Many novel drug delivery technologies have been investigated to bypass or disrupt the BBB/BBTB, including convection enhanced delivery, focused ultrasound, nanoparticle-mediated delivery, and intranasal delivery, all of which are yet to be clinically established for the treatment of DIPG. Herein, we review what is known about the BBB/BBTB and discuss the current status, limitations, and advances of conventional and novel treatments to improving brain drug delivery in DIPG.


Subject(s)
Antineoplastic Agents , Blood-Brain Barrier , Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Drug Delivery Systems , Humans , Brain Stem Neoplasms/drug therapy , Animals , Diffuse Intrinsic Pontine Glioma/drug therapy , Drug Delivery Systems/methods , Blood-Brain Barrier/metabolism , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacokinetics , Glioma/drug therapy
2.
Neurosurg Rev ; 47(1): 212, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38727935

ABSTRACT

We aimed to evaluate the relationship between imaging features, therapeutic responses (comparative cross-product and volumetric measurements), and overall survival (OS) in pediatric diffuse intrinsic pontine glioma (DIPG). A total of 134 patients (≤ 18 years) diagnosed with DIPG were included. Univariate and multivariate analyses were performed to evaluate correlations of clinical and imaging features and therapeutic responses with OS. The correlation between cross-product (CP) and volume thresholds in partial response (PR) was evaluated by linear regression. The log-rank test was used to compare OS patients with discordant therapeutic response classifications and those with concordant classifications. In univariate analysis, characteristics related to worse OS included lower Karnofsky, larger extrapontine extension, ring-enhancement, necrosis, non-PR, and increased ring enhancement post-radiotherapy. In the multivariate analysis, Karnofsky, necrosis, extrapontine extension, and therapeutic response can predict OS. A 25% CP reduction (PR) correlated with a 32% volume reduction (R2 = 0.888). Eight patients had discordant therapeutic response classifications according to CP (25%) and volume (32%). This eight patients' median survival time was 13.0 months, significantly higher than that in the non-PR group (8.9 months), in which responses were consistently classified as non-PR based on CP (25%) and volume (32%). We identified correlations between imaging features, therapeutic responses, and OS; this information is crucial for future clinical trials. Tumor volume may represent the DIPG growth pattern more accurately than CP measurement and can be used to evaluate therapeutic response.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Humans , Brain Stem Neoplasms/diagnostic imaging , Brain Stem Neoplasms/therapy , Brain Stem Neoplasms/mortality , Brain Stem Neoplasms/pathology , Male , Child , Female , Adolescent , Diffuse Intrinsic Pontine Glioma/therapy , Child, Preschool , Treatment Outcome , Magnetic Resonance Imaging , Infant , Retrospective Studies , Glioma/therapy , Glioma/pathology , Glioma/diagnostic imaging , Glioma/mortality
3.
Anticancer Res ; 44(6): 2325-2333, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38821589

ABSTRACT

BACKGROUND/AIM: In the past decade, diffuse intrinsic pontine glioma (DIPG), the most common childhood brainstem glioma, has benefitted from an increase in tissue-based research because of improved biopsy collection techniques. However, the adaptive immune receptor (IR) features represented by tumor material and tumor infiltrating lymphocytes have remained poorly understood. MATERIALS AND METHODS: Herein, we characterized the adaptive immune parameters of DIPG through the recovery of IR recombination reads from RNAseq files representing initial and progressive DIPG samples. RESULTS: An elevated level of immunoglobulin gene expression in the progressive DIPG sample files and a reduced number of bacterial sequencing read recoveries in comparison to RNAseq files representing the initial form of DIPG, was found. Furthermore, the RNAseq files representing both initial and progressive DIPG samples had significant numbers of reads representing Cutibacterium acnes, a bacterium previously linked to prostate cancer development. Results also indicated an opportunity to distinguish overall survival probabilities based on IGL complementarity determining region-3 amino acid sequence physicochemical parameters. CONCLUSION: Genomics analyses allow for a better understanding of adaptive IR features and bacterial infections in the DIPG setting.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Humans , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/microbiology , Brain Stem Neoplasms/pathology , Diffuse Intrinsic Pontine Glioma/genetics , Diffuse Intrinsic Pontine Glioma/microbiology , Diffuse Intrinsic Pontine Glioma/pathology , Male , Disease Progression , Child , Immunoglobulins/genetics , Female , Child, Preschool , Lymphocytes, Tumor-Infiltrating/immunology
4.
Med J Aust ; 220(10): 533-538, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38699949

ABSTRACT

INTRODUCTION: The main mission of the Australian and New Zealand Children's Haematology and Oncology Group (ANZCHOG) is to develop and facilitate local access to the world's leading evidence-based clinical trials for all paediatric cancers, including brain tumours, as soon as practically possible. Diffuse intrinsic pontine gliomas (DIPGs) - a subset of a larger group of tumours now termed diffuse midline glioma, H3K27-altered (DMG) - are paediatric brain cancers with less than 10% survival at two years. In the absence of any proven curative therapies, significant recent advancements have been made in pre-clinical and clinical research, leading many to seek integration of novel therapies early into standard practice. Despite these innovative therapeutic approaches, DIPG remains an incurable disease for which novel surgical, imaging, diagnostic, radiation and systemic therapy approaches are needed. MAIN RECOMMENDATIONS: All patients with DIPG should be discussed in multidisciplinary neuro-oncology meetings (including pathologists, neuroradiologists, radiation oncologists, neurosurgeons, medical oncologists) at diagnosis and at relapse or progression. Radiation therapy to the involved field remains the local and international standard of care treatment. Proton therapy does not yield a superior survival outcome compared with photon therapy and patients should undergo radiation therapy with the available modality (photon or proton) at their treatment centre. Patients may receive concurrent chemotherapy or radiation-sensitising agents as part of a clinical trial. Biopsy should be offered to facilitate consideration of experimental therapies and eligibility for clinical trial participation. After radiation therapy, each patient should be managed individually with either observation or considered for enrolment on a clinical trial, if eligible, after full discussion with the family. Re-irradiation can be considered for progressive disease. CHANGES IN MANAGEMENT AS A RESULT OF THE GUIDELINE: Every child diagnosed with DIPG should be offered enrolment on a clinical trial where available. Access to investigational drugs without biological rationale outside the clinical trial setting is not supported. In case of potentially actionable target identification with molecular profiling and absence of a suitable clinical trial, rational targeted therapies can be considered through compassionate access programs.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Humans , New Zealand , Brain Stem Neoplasms/therapy , Brain Stem Neoplasms/diagnosis , Australia , Child , Diffuse Intrinsic Pontine Glioma/therapy , Diffuse Intrinsic Pontine Glioma/diagnosis
5.
Acta Neuropathol Commun ; 12(1): 71, 2024 05 05.
Article in English | MEDLINE | ID: mdl-38706008

ABSTRACT

Diffuse Intrinsic Pontine Glioma (DIPG) is a highly aggressive and fatal pediatric brain cancer. One pre-requisite for tumor cells to infiltrate is adhesion to extracellular matrix (ECM) components. However, it remains largely unknown which ECM proteins are critical in enabling DIPG adhesion and migration and which integrin receptors mediate these processes. Here, we identify laminin as a key ECM protein that supports robust DIPG cell adhesion and migration. To study DIPG infiltration, we developed a DIPG-neural assembloid model, which is composed of a DIPG spheroid fused to a human induced pluripotent stem cell-derived neural organoid. Using this assembloid model, we demonstrate that knockdown of laminin-associated integrins significantly impedes DIPG infiltration. Moreover, laminin-associated integrin knockdown improves DIPG response to radiation and HDAC inhibitor treatment within the DIPG-neural assembloids. These findings reveal the critical role of laminin-associated integrins in mediating DIPG progression and drug response. The results also provide evidence that disrupting integrin receptors may offer a novel therapeutic strategy to enhance DIPG treatment outcomes. Finally, these results establish DIPG-neural assembloid models as a powerful tool to study DIPG disease progression and enable drug discovery.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Integrins , Laminin , Humans , Laminin/metabolism , Integrins/metabolism , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/pathology , Brain Stem Neoplasms/metabolism , Brain Stem Neoplasms/therapy , Diffuse Intrinsic Pontine Glioma/pathology , Diffuse Intrinsic Pontine Glioma/genetics , Cell Adhesion/drug effects , Cell Movement , Cell Line, Tumor , Glioma/pathology , Glioma/metabolism , Glioma/genetics , Glioma/therapy
6.
World Neurosurg ; 187: e870-e882, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38734176

ABSTRACT

BACKGROUND: Diffuse intrinsic pontine gliomas are aggressive tumors that carry a poor prognosis with a 2-year survival rate of <10%. The imaging appearance is often pathognomonic, and surgical biopsy is not mandatory to initiate treatment in children. Studies of biopsy samples provide insight into the disease's molecular pathobiology and open prospects for targeted therapy. This study was conducted to determine the diagnostic yield and safety of stereotactic biopsies. METHODS: This is a prospective observational study from a single tertiary health care center. All patients with clinical and radiological features diagnostic of diffuse intrinsic pontine gliomas (DIPGs) who underwent biopsy from July 2018 to June 2023 were included. Biopsies were performed using either stereotactic frame-based, frameless, or endoscopic techniques. RESULTS: A total of 165 patients with DIPGs were evaluated in the study period. The option of biopsy with its associated risks and benefits was offered to all patients. A total of 76 biopsies were performed in 74 patients (40 children and 34 adults, including 2 repeat biopsies). The median age was 15 years. Diffuse midline gliomas, H3K27M altered, was the most common histopathological diagnosis (85% pediatric and 55.9% adults). The diagnostic efficacy of the procedure was 94.7%. The complication rate was 10.8%, with no permanent neurological deficits due to surgery. There was no procedure-related mortality. CONCLUSIONS: Establishing the safety of the procedure could be an important step toward popularizing the concept, which might offer a better understanding of the disease. Brainstem eloquence and a lack of direct benefit to patients are the primary obstacles to brainstem biopsy.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Humans , Male , Female , Adolescent , Child , Brain Stem Neoplasms/pathology , Brain Stem Neoplasms/surgery , Brain Stem Neoplasms/diagnostic imaging , Adult , Prospective Studies , Biopsy/methods , Biopsy/adverse effects , Young Adult , Diffuse Intrinsic Pontine Glioma/pathology , Child, Preschool , Middle Aged , Stereotaxic Techniques/adverse effects , Pons/pathology
7.
Cancer Lett ; 590: 216876, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38609002

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) is a childhood malignancy of the brainstem with a dismal prognosis. Despite recent advances in its understanding at the molecular level, the prognosis of DIPG has remained unchanged. This article aims to review the current understanding of the genetic pathophysiology of DIPG and to highlight promising therapeutic targets. Various DIPG treatment strategies have been investigated in pre-clinical studies, several of which have shown promise and have been subsequently translated into ongoing clinical trials. Ultimately, a multifaceted therapeutic approach that targets cell-intrinsic alterations, the micro-environment, and augments the immune system will likely be necessary to eradicate DIPG.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Humans , Diffuse Intrinsic Pontine Glioma/genetics , Diffuse Intrinsic Pontine Glioma/therapy , Diffuse Intrinsic Pontine Glioma/pathology , Diffuse Intrinsic Pontine Glioma/drug therapy , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/therapy , Brain Stem Neoplasms/pathology , Brain Stem Neoplasms/drug therapy , Prognosis , Tumor Microenvironment , Molecular Targeted Therapy/methods
8.
J Pediatr Hematol Oncol ; 46(4): 211-215, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38573000

ABSTRACT

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.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Humans , Female , Brain Stem Neoplasms/pathology , Brain Stem Neoplasms/therapy , Brain Stem Neoplasms/mortality , Diffuse Intrinsic Pontine Glioma/pathology , Diffuse Intrinsic Pontine Glioma/therapy , Diffuse Intrinsic Pontine Glioma/genetics , Child , Survivorship , Cancer Survivors , Fatal Outcome , Isocitrate Dehydrogenase/genetics , Prognosis , Mutation
9.
Pediatr Blood Cancer ; 71(5): e30929, 2024 May.
Article in English | MEDLINE | ID: mdl-38430472

ABSTRACT

BACKGROUND: Re-irradiation (reRT) increases survival in locally recurrent diffuse intrinsic pontine glioma (DIPG). There is no standard dose and fractionation for reRT, but conventional fractionation (CF) is typically used. We report our institutional experience of reRT for DIPG, which includes hypofractionation (HF). METHODS: We reviewed pediatric patients treated with brainstem reRT for DIPG at our institution from 2012 to 2022. Patients were grouped by HF or CF. Outcomes included steroid use, and overall survival (OS) was measured from both diagnosis and start of reRT. RESULTS: Of 22 patients who received reRT for DIPG, two did not complete their course due to clinical decline. Of the 20 who completed reRT, the dose was 20-30 Gy in 2-Gy fractions (n = 6) and 30-36 Gy in 3-Gy fractions (n = 14). Median age was 5 years (range: 3-14), median interval since initial RT was 8 months (range: 3-20), and 12 received concurrent bevacizumab. Median OS from diagnosis was 18 months [95% confidence interval: 17-24]. Median OS from start of reRT for HF versus CF was 8.2 and 7.5 months, respectively (p = .20). Thirteen (93%) in the HF group and three (75%) in the CF group tapered pre-treatment steroid dose down or off within 2 months after reRT due to clinical improvement. There was no significant difference in steroid taper between HF and CF (p = .4). No patients developed radionecrosis. CONCLUSION: reRT with HF achieved survival duration comparable to published outcomes and effectively palliated symptoms. Future investigation of this regimen in the context of new systemic therapies and upfront HF is warranted.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Re-Irradiation , Adolescent , Child , Child, Preschool , Humans , Brain Stem Neoplasms/radiotherapy , Diffuse Intrinsic Pontine Glioma/radiotherapy , Radiation Dose Hypofractionation , Steroids
10.
Childs Nerv Syst ; 40(6): 1671-1680, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38478066

ABSTRACT

PURPOSE: Pediatric diffuse intrinsic pontine glioma (DIPG) is a fatal disease associated with a median survival of < 1 year despite aggressive treatments. This retrospective study analyzed the treatment outcomes of patients aged < 18 years who were diagnosed with DIPG between 2012 and 2022 and who received different chemotherapy regimens. METHODS: After radiotherapy, patients with DIPG received nimotuzumab-vinorelbine combination or temozolomide-containing therapy. When nimotuzumab was unavailable, it was replaced by vincristine, etoposide, and carboplatin/cyclophosphamide (VECC). Temozolomide was administered as a single agent or a part of the combination chemotherapy comprising temozolomide, irinotecan, and bevacizumab. Furthermore, 1- and 3-year overall survival (OS), progression-free survival (PFS), and median OS and PFS were analyzed. RESULTS: The median age of 40 patients with DIPG was 97 ± 46.93 (23-213) months; the median follow-up time was 12 months. One and 3-year OS were 35.0% and 7.5%, respectively. Median OS was 12 months in all patients (n = 40), and it was 16, 10, and 11 months in those who received first-line nimotuzumab-vinorelbine combination (n = 13), temozolomide-based (n = 14), and VECC (n = 6) chemotherapy regimens, respectively (p = 0.360). One patient who received gefitinib survived for 16 months. Conversely, patients who never received radiotherapy and any antineoplastic medicamentous therapy (n = 6) had a median OS of 4 months. CONCLUSION: Nimotuzumab-vinorelbine combination therapy prolonged OS by 6 months compared with temozolomide-containing chemotherapy, although the difference was not statistically significant.


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols , Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Humans , Female , Child , Male , Brain Stem Neoplasms/drug therapy , Child, Preschool , Retrospective Studies , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Adolescent , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , Diffuse Intrinsic Pontine Glioma/drug therapy , Temozolomide/therapeutic use , Temozolomide/administration & dosage , Vinblastine/administration & dosage , Vinblastine/therapeutic use , Vinblastine/analogs & derivatives , Infant , Treatment Outcome
11.
J Neurosurg Pediatr ; 33(6): 507-515, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38489807

ABSTRACT

OBJECTIVE: Given the lack of a definitive treatment and the poor prognosis of patients with diffuse midline glioma (DMG) and diffuse intrinsic pontine glioma (DIPG), socioeconomic status (SES) may affect treatment access and therefore survival. Therefore, this study aimed to examine the relationship between SES and treatment modalities, progression-free survival (PFS), and overall survival (OS) in children with DMG/DIPG. METHODS: A retrospective, single-institution review was conducted of medical records of patients ≤ 18 years of age who had DMG or DIPG that was diagnosed between 2000 and 2022. Patient demographics, surgical interventions, chemotherapy, radiation therapy, clinical trial enrollment, and medical care-related travel were extracted. SES variables (education and mean income) for associated patient census tracts were collected and stratified. Statistical analysis using unpaired t-tests, chi-square analysis, and log-rank tests was conducted. RESULTS: Of the 96 patients who met the inclusion criteria, the majority were female (59%) and non-Hispanic White (57%). The median PFS, median OS, and time from diagnosis to treatment did not differ between races/ethnicities or sex. Ninety-one of 96 patients had census tract data available. Patients from higher-income census tracts (> 50% of families with annual household income greater than $50,000) had a longer median OS (480 vs 235 days, p < 0.001) and traveled significantly longer distances for medical care (1550 vs 1114 miles, p = 0.048) than families from lower-income census tracts. Patients from the highest education quartile traveled significantly farther for treatment than the lowest education quartile (mean 2964 vs 478 miles, p = 0.047). Patients who received both oral and intravenous chemotherapy were more likely to be from higher-income census tracts than those who received intravenous or no chemotherapy. Duration of PFS, rates of clinical trial enrollment, biopsy rates, H3K27 mutation status, ventriculoperitoneal shunt placement rates, and radiotherapy rates were not associated with SES variables. CONCLUSIONS: Patients from families from higher-income census tracts experienced longer OS and traveled farther for treatment. Patients from families from higher-education-level census tracts traveled more often for treatment. The authors' findings suggest that SES influences DMG and DIPG OS. More studies should be done to understand the role of SES in the outcomes of children with DMG/DIPG.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Glioma , Social Class , Humans , Female , Male , Brain Stem Neoplasms/therapy , Brain Stem Neoplasms/pathology , Child , Retrospective Studies , Diffuse Intrinsic Pontine Glioma/therapy , Child, Preschool , Glioma/therapy , Glioma/pathology , Glioma/mortality , Adolescent , Treatment Outcome , Infant , Progression-Free Survival
12.
BMJ Open ; 14(3): e077613, 2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38503417

ABSTRACT

INTRODUCTION: Diffuse intrinsic pontine glioma (DIPG) and paediatric high-grade glioma (pHGG) are aggressive glial tumours, for which conventional treatment modalities fall short. Dendritic cell (DC)-based immunotherapy is being investigated as a promising and safe adjuvant therapy. The Wilms' tumour protein (WT1) is a potent target for this type of antigen-specific immunotherapy and is overexpressed in DIPG and pHGG. Based on this, we designed a non-randomised phase I/II trial, assessing the feasibility and safety of WT1 mRNA-loaded DC (WT1/DC) immunotherapy in combination with conventional treatment in pHGG and DIPG. METHODS AND ANALYSIS: 10 paediatric patients with newly diagnosed or pretreated HGG or DIPG were treated according to the trial protocol. The trial protocol consists of leukapheresis of mononuclear cells, the manufacturing of autologous WT1/DC vaccines and the combination of WT1/DC-vaccine immunotherapy with conventional antiglioma treatment. In newly diagnosed patients, this comprises chemoradiation (oral temozolomide 90 mg/m2 daily+radiotherapy 54 Gy in 1.8 Gy fractions) followed by three induction WT1/DC vaccines (8-10×106 cells/vaccine) given on a weekly basis and a chemoimmunotherapy booster phase consisting of six 28-day cycles of oral temozolomide (150-200 mg/m2 on days 1-5) and a WT1/DC vaccine on day 21. In pretreated patients, the induction and booster phase are combined with best possible antiglioma treatment at hand. Primary objectives are to assess the feasibility of the production of mRNA-electroporated WT1/DC vaccines in this patient population and to assess the safety and feasibility of combining conventional antiglioma treatment with the proposed immunotherapy. Secondary objectives are to investigate in vivo immunogenicity of WT1/DC vaccination and to assess disease-specific and general quality of life. ETHICS AND DISSEMINATION: The ethics committee of the Antwerp University Hospital and the University of Antwerp granted ethics approval. Results of the clinical trial will be shared through publication in a peer-reviewed journal and presentations at conferences. TRIAL REGISTRATION NUMBER: NCT04911621.


Subject(s)
Cancer Vaccines , Diffuse Intrinsic Pontine Glioma , Glioma , Kidney Neoplasms , Vaccines , Wilms Tumor , Humans , Child , WT1 Proteins/metabolism , Temozolomide/therapeutic use , Diffuse Intrinsic Pontine Glioma/metabolism , Belgium , Quality of Life , Glioma/therapy , Glioma/pathology , Wilms Tumor/metabolism , Immunotherapy/methods , Dendritic Cells , RNA, Messenger , Cancer Vaccines/therapeutic use , Clinical Trials, Phase II as Topic , Clinical Trials, Phase I as Topic
13.
Int J Mol Sci ; 25(6)2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38542334

ABSTRACT

The BMP pathway is one of the major signaling pathways in embryonic development, ontogeny and homeostasis, identified many years ago by pioneers in developmental biology. Evidence of the deregulation of its activity has also emerged in many cancers, with complex and sometimes opposing effects. Recently, its role has been suspected in Diffuse Midline Gliomas (DMG), among which Diffuse Intrinsic Pontine Gliomas (DIPG) are one of the most complex challenges in pediatric oncology. Genomic sequencing has led to understanding part of their molecular etiology, with the identification of histone H3 mutations in a large proportion of patients. The epigenetic remodeling associated with these genetic alterations has also been precisely described, creating a permissive context for oncogenic transcriptional program activation. This review aims to describe the new findings about the involvement of BMP pathway activation in these tumors, placing their appearance in a developmental context. Targeting the oncogenic synergy resulting from this pathway activation in an H3K27M context could offer new therapeutic perspectives based on targeting treatment-resistant cell states.


Subject(s)
Diffuse Intrinsic Pontine Glioma , Glioma , Humans , Child , Glioma/metabolism , Histones/metabolism , Diffuse Intrinsic Pontine Glioma/genetics , Diffuse Intrinsic Pontine Glioma/metabolism , Diffuse Intrinsic Pontine Glioma/pathology , Mutation , Signal Transduction , Bone Morphogenetic Proteins/metabolism
14.
Cell Rep ; 43(2): 113707, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38306270

ABSTRACT

Diffuse intrinsic pontine gliomas (DIPGs) are deadly pediatric brain tumors, non-resectable due to brainstem localization and diffusive growth. Over 80% of DIPGs harbor a mutation in histone 3 (H3.3 or H3.1) resulting in a lysine-to-methionine substitution (H3K27M). Patients with DIPG have a dismal prognosis with no effective therapy. We show that histone deacetylase (HDAC) inhibitors lead to a significant reduction in the H3.3K27M protein (up to 80%) in multiple glioma cell lines. We discover that the SB939-mediated H3.3K27M loss is partially blocked by a lysosomal inhibitor, chloroquine. The H3.3K27M loss is facilitated by co-occurrence of H2A.Z, as evidenced by the knockdown of H2A.Z isoforms. Chromatin immunoprecipitation sequencing (ChIP-seq) analysis confirms the occupancy of H3.3K27M and H2A.Z at the same SB939-inducible genes. We discover a mechanism showing that HDAC inhibition in DIPG leads to pharmacological modulation of the oncogenic H3.3K27M protein levels. These findings show the possibility of directly targeting the H3.3K27M oncohistone.


Subject(s)
Brain Neoplasms , Diffuse Intrinsic Pontine Glioma , Glioma , Humans , Child , Histones , Mutant Proteins , Glioma/genetics , Brain Neoplasms/genetics , Histone Deacetylase Inhibitors/pharmacology
15.
J Clin Invest ; 134(6)2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38319732

ABSTRACT

Diffuse midline glioma (DMG), including tumors diagnosed in the brainstem (diffuse intrinsic pontine glioma; DIPG), are uniformly fatal brain tumors that lack effective treatment. Analysis of CRISPR/Cas9 loss-of-function gene deletion screens identified PIK3CA and MTOR as targetable molecular dependencies across patient derived models of DIPG, highlighting the therapeutic potential of the blood-brain barrier-penetrant PI3K/Akt/mTOR inhibitor, paxalisib. At the human-equivalent maximum tolerated dose, mice treated with paxalisib experienced systemic glucose feedback and increased insulin levels commensurate with patients using PI3K inhibitors. To exploit genetic dependence and overcome resistance while maintaining compliance and therapeutic benefit, we combined paxalisib with the antihyperglycemic drug metformin. Metformin restored glucose homeostasis and decreased phosphorylation of the insulin receptor in vivo, a common mechanism of PI3K-inhibitor resistance, extending survival of orthotopic models. DIPG models treated with paxalisib increased calcium-activated PKC signaling. The brain penetrant PKC inhibitor enzastaurin, in combination with paxalisib, synergistically extended the survival of multiple orthotopic patient-derived and immunocompetent syngeneic allograft models; benefits potentiated in combination with metformin and standard-of-care radiotherapy. Therapeutic adaptation was assessed using spatial transcriptomics and ATAC-Seq, identifying changes in myelination and tumor immune microenvironment crosstalk. Collectively, this study has identified what we believe to be a clinically relevant DIPG therapeutic combinational strategy.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Glioma , Metformin , Humans , Mice , Animals , Diffuse Intrinsic Pontine Glioma/drug therapy , Diffuse Intrinsic Pontine Glioma/genetics , Phosphatidylinositol 3-Kinases/genetics , Brain Stem Neoplasms/drug therapy , Brain Stem Neoplasms/genetics , Glioma/drug therapy , Glioma/genetics , Glioma/pathology , TOR Serine-Threonine Kinases/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Glucose , Metformin/pharmacology , Tumor Microenvironment
16.
Cancer Res ; 84(4): 598-615, 2024 02 15.
Article in English | MEDLINE | ID: mdl-38095539

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) is the most aggressive pediatric brain tumor, and the oncohistone H3.3K27M mutation is associated with significantly worse clinical outcomes. Despite extensive research efforts, effective approaches for treating DIPG are lacking. Through drug screening, we identified the combination of gemcitabine and fimepinostat as a potent therapeutic intervention for H3.3K27M DIPG. H3.3K27M facilitated gemcitabine-induced apoptosis in DIPG, and gemcitabine stabilized and activated p53, including increasing chromatin accessibility for p53 at apoptosis-related loci. Gemcitabine simultaneously induced a prosurvival program in DIPG through activation of RELB-mediated NF-κB signaling. Specifically, gemcitabine induced the transcription of long terminal repeat elements, activated cGAS-STING signaling, and stimulated noncanonical NF-κB signaling. A drug screen in gemcitabine-treated DIPG cells revealed that fimepinostat, a dual inhibitor of HDAC and PI3K, effectively suppressed the gemcitabine-induced NF-κB signaling in addition to blocking PI3K/AKT activation. Combination therapy comprising gemcitabine and fimepinostat elicited synergistic antitumor effects in vitro and in orthotopic H3.3K27M DIPG xenograft models. Collectively, p53 activation using gemcitabine and suppression of RELB-mediated NF-κB activation and PI3K/AKT signaling using fimepinostat is a potential therapeutic strategy for treating H3.3K27M DIPG. SIGNIFICANCE: Gemcitabine activates p53 and induces apoptosis to elicit antitumor effects in H3.3K27M DIPG, which can be enhanced by blocking NF-κB and PI3K/AKT signaling with fimepinostat, providing a synergistic combination therapy for DIPG.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Morpholines , Pyrimidines , Sulfur Compounds , Child , Humans , Diffuse Intrinsic Pontine Glioma/genetics , Gemcitabine , NF-kappa B , Brain Stem Neoplasms/drug therapy , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/pathology , Proto-Oncogene Proteins c-akt , Phosphatidylinositol 3-Kinases , Tumor Suppressor Protein p53
17.
Neuro Oncol ; 26(4): 735-748, 2024 04 05.
Article in English | MEDLINE | ID: mdl-38011799

ABSTRACT

BACKGROUND: Diffuse intrinsic pontine gliomas (DIPG/DMG) are devastating pediatric brain tumors with extraordinarily limited treatment options and uniformly fatal prognosis. Histone H3K27M mutation is a common recurrent alteration in DIPG and disrupts epigenetic regulation. We hypothesize that genome-wide H3K27M-induced epigenetic dysregulation makes tumors vulnerable to epigenetic targeting. METHODS: We performed a screen of compounds targeting epigenetic enzymes to identify potential inhibitors for the growth of patient-derived DIPG cells. We further carried out transcriptomic and genomic landscape profiling including RNA-seq and CUT&RUN-seq as well as shRNA-mediated knockdown to assess the effects of chaetocin and SUV39H1, a target of chaetocin, on DIPG growth. RESULTS: High-throughput small-molecule screening identified an epigenetic compound chaetocin as a potent blocker of DIPG cell growth. Chaetocin treatment selectively decreased proliferation and increased apoptosis of DIPG cells and significantly extended survival in DIPG xenograft models, while restoring H3K27me3 levels. Moreover, the loss of H3K9 methyltransferase SUV39H1 inhibited DIPG cell growth. Transcriptomic and epigenomic profiling indicated that SUV39H1 loss or inhibition led to the downregulation of stemness and oncogenic networks including growth factor receptor signaling and stemness-related programs; however, D2 dopamine receptor (DRD2) signaling adaptively underwent compensatory upregulation conferring resistance. Consistently, a combination of chaetocin treatment with a DRD2 antagonist ONC201 synergistically increased the antitumor efficacy. CONCLUSIONS: Our studies reveal a therapeutic vulnerability of DIPG cells through targeting the SUV39H1-H3K9me3 pathway and compensatory signaling loops for treating this devastating disease. Combining SUV39H1-targeting chaetocin with other agents such as ONC201 may offer a new strategy for effective DIPG treatment.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Imidazoles , Pyridines , Pyrimidines , Child , Humans , Epigenesis, Genetic , Histones/genetics , Diffuse Intrinsic Pontine Glioma/genetics , Brain Stem Neoplasms/drug therapy , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/pathology , Methyltransferases/genetics , Methyltransferases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Piperazines
18.
Mol Cancer Ther ; 23(1): 24-34, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-37723046

ABSTRACT

Therapeutic resistance remains a major obstacle to successful clinical management of diffuse intrinsic pontine glioma (DIPG), a high-grade pediatric tumor of the brain stem. In nearly all patients, available therapies fail to prevent progression. Innovative combinatorial therapies that penetrate the blood-brain barrier and lead to long-term control of tumor growth are desperately needed. We identified mechanisms of resistance to radiotherapy, the standard of care for DIPG. On the basis of these findings, we rationally designed a brain-penetrant small molecule, MTX-241F, that is a highly selective inhibitor of EGFR and PI3 kinase family members, including the DNA repair protein DNA-PK. Preliminary studies demonstrated that micromolar levels of this inhibitor can be achieved in murine brain tissue and that MTX-241F exhibits promising single-agent efficacy and radiosensitizing activity in patient-derived DIPG neurospheres. Its physiochemical properties include high exposure in the brain, indicating excellent brain penetrance. Because radiotherapy results in double-strand breaks that are repaired by homologous recombination (HR) and non-homologous DNA end joining (NHEJ), we have tested the combination of MTX-241F with an inhibitor of Ataxia Telangiectasia Mutated to achieve blockade of HR and NHEJ, respectively, with or without radiotherapy. When HR blockers were combined with MTX-241F and radiotherapy, synthetic lethality was observed, providing impetus to explore this combination in clinically relevant models of DIPG. Our data provide proof-of-concept evidence to support advanced development of MTX-241F for the treatment of DIPG. Future studies will be designed to inform rapid clinical translation to ultimately impact patients diagnosed with this devastating disease.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Humans , Child , Mice , Animals , Diffuse Intrinsic Pontine Glioma/drug therapy , Diffuse Intrinsic Pontine Glioma/genetics , Diffuse Intrinsic Pontine Glioma/metabolism , Neoplasm Recurrence, Local , DNA Repair , Signal Transduction , DNA/therapeutic use , Brain Stem Neoplasms/drug therapy , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/pathology
19.
Neuro Oncol ; 26(Supplement_2): S110-S124, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38102230

ABSTRACT

H3 K27M-mutant diffuse glioma is a recently identified brain tumor associated with poor prognosis. As of 2016, it is classified by the World Health Organization as a distinct form of grade IV glioma. Despite recognition as an important prognostic and diagnostic feature in diffuse glioma, radiation remains the sole standard of care and no effective systemic therapies are available for H3K27M mutant tumors. This review will detail treatment interventions applied to diffuse midline glioma and diffuse intrinsic pontine glioma (DIPG) prior to the identification of the H3 K27M mutation, the current standard-of-care for H3 K27M-mutant diffuse glioma treatment, and ongoing clinical trials listed on www.clinicaltrials.gov evaluating novel therapeutics in this population. Current clinical trials were identified using clinicaltrials.gov, and studies qualifying for this analysis were active or ongoing interventional trials that evaluated a therapy in at least 1 treatment arm or cohort comprised exclusively of patients with DIPG and H3 K27M-mutant glioma. Forty-one studies met these criteria, including trials evaluating H3 K27M vaccination, chimeric antigen receptor T-cell therapy, and small molecule inhibitors. Ongoing evaluation of novel therapeutics is necessary to identify safe and effective interventions in this underserved patient population.


Subject(s)
Diffuse Intrinsic Pontine Glioma , Glioma , Histones , Mutation , Humans , Brain Neoplasms/therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Stem Neoplasms/therapy , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/pathology , Diffuse Intrinsic Pontine Glioma/therapy , Diffuse Intrinsic Pontine Glioma/genetics , Diffuse Intrinsic Pontine Glioma/pathology , Glioma/genetics , Glioma/therapy , Glioma/pathology , Histones/genetics , Prognosis
20.
Rio de Janeiro; s.n; 2024. 23 p.
Thesis in Portuguese | Coleciona SUS | ID: biblio-1537925

ABSTRACT

O glioma pontino intrínseco difuso (GPID) é um tumor cerebral maligno e raro em crianças, apresentando baixa sobrevida. Este estudo tem como objetivo analisar e agrupar estudos sobre o glioma pontino intrínseco difuso, explorando métodos diagnósticos, tratamentos e desafios associados. Trata-se de uma revisão integrada da literatura, com pesquisa bibliográfica em bases de dados PubMed, Google Scholar, Springer link, Willey Online Library, em trabalhos publicados dentro de um período de sete anos (2017-2023). Essa revisão abrange 341 pacientes com GPID, com o uso de métodos como ressonância magnética (RNM), biópsias e diversos tratamentos (Zr-bevacizumabe, células T GD2-CAR, radioterapia, vorinostat). A classificação em sobreviventes de curto e longo prazos, heterogeneidade na captação de Zr-bevacizumabe e interações farmacocinéticas são discutidas, assim como os desafios da radioterapia paliativa. A idade e a duração dos sintomas afetam a sobrevida. As restrições em procedimentos invasivos, como biópsias, ilustram a complexidade no diagnóstico e tratamento do GPID, acentuando desafios clínicos. É possível concluir que existe necessidade de mais pesquisas para desenvolver intervenções inovadoras e precisas, no intuito de melhorar o prognóstico associado ao GPID


Diffuse intrinsic pontine glioma (DIPG) is a malignant and rare brain tumor in children, with poor survival. This study aims to analyze and group studies on diffuse intrinsic pontine glioma, exploring diagnostic methods, treatments and associated challenges. This is an integrated literature review, with bibliographic research in databases PubMed, Google Scholar, Springer link, Willey Online Library, in works published within a period of seven years (2017-2023). This review covers 341 patients with IDPG, using methods such as magnetic resonance imaging (MRI), biopsies and various treatments (Zr-bevacizumab, GD2-CAR T cells, radiotherapy, vorinostat). Classification into short- and longterm survivors, heterogeneity in Zr-bevacizumab uptake, and pharmacokinetic interactions are discussed, as are the challenges of palliative radiotherapy. Age and duration of symptoms affect survival. Restrictions on invasive procedures, such as biopsies, illustrate the complexity in diagnosing and treating IDPG, accentuating clinical challenges. It is possible to conclude that there is a need for more research to develop innovative and precise interventions, with the aim of improving the prognosis associated with IDPG


Subject(s)
Humans , Male , Female , Therapeutics , Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma
SELECTION OF CITATIONS
SEARCH DETAIL
...