Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.954
Filter
1.
Curr Radiopharm ; 17(1): 55-67, 2024.
Article in English | MEDLINE | ID: mdl-38817005

ABSTRACT

BACKGROUND: Exposure to physical contamination during chemotherapy, including non-ionizing electromagnetic fields, raises concerns about the widespread sources of exposure to this type of radiation. Glioblastoma multiforme (GBM) is an aggressive central nervous system tumor that is hard to treat due to resistance to drugs such as temozolomide (TMZ). OBJECTIVE: Electromagnetic fields (EMF) and haloperidol (HLP) may have anticancer effects. In this study, we investigated the effects of TMZ, HLP, and EMF on GBM cell lines and analyzed the association between non-ionizing radiation and the risk of change in drug performance. METHODS: Cell viability and reactive oxygen species (ROS) generation were measured by MTT and NBT assay, respectively. Then, the expression levels of breast cancer-resistant protein (BCRP), Bax, Bcl2, Nestin, vascular endothelial growth factor (VEGF) genes, and P53, Bax, and Bcl2 Proteins were evaluated by real-time PCR and western blot. RESULTS: Co-treatment of GBM cells by HLP and TMZ enhanced apoptosis in T-98G and A172 cells by increasing the expression of P53 and Bax and decreasing Bcl-2. Interestingly, exposure of GBM cells to EMF decreased apoptosis in the TMZ+HLP group. CONCLUSION: In conclusion, EMF reduced the synergistic effect of TMZ and HLP. This hypothesis that patients who are treated for brain tumors and suffer from depression should not be exposed to EMF is proposed in the present study. There appears to be an urgent need to reconsider exposure limits for low-frequency magnetic fields, based on experimental and epidemiological research, the relationship between exposure to non-ionizing radiation and adverse human health effects.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2 , Apoptosis , Cell Survival , Electromagnetic Fields , Haloperidol , Neoplasm Proteins , Nestin , Temozolomide , Vascular Endothelial Growth Factor A , Humans , Apoptosis/drug effects , Apoptosis/radiation effects , Nestin/metabolism , Temozolomide/pharmacology , Haloperidol/pharmacology , Vascular Endothelial Growth Factor A/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Neoplasm Proteins/metabolism , Neoplasm Proteins/biosynthesis , Glioblastoma/radiotherapy , Glioblastoma/metabolism , Reactive Oxygen Species/metabolism , Brain Neoplasms/radiotherapy , Brain Neoplasms/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Glioma/radiotherapy , Glioma/metabolism , Glioma/pathology
2.
Nat Commun ; 15(1): 4210, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806504

ABSTRACT

The chemokine CXCL12 promotes glioblastoma (GBM) recurrence after radiotherapy (RT) by facilitating vasculogenesis. Here we report outcomes of the dose-escalation part of GLORIA (NCT04121455), a phase I/II trial combining RT and the CXCL12-neutralizing aptamer olaptesed pegol (NOX-A12; 200/400/600 mg per week) in patients with incompletely resected, newly-diagnosed GBM lacking MGMT methylation. The primary endpoint was safety, secondary endpoints included maximum tolerable dose (MTD), recommended phase II dose (RP2D), NOX-A12 plasma levels, topography of recurrence, tumor vascularization, neurologic assessment in neuro-oncology (NANO), quality of life (QOL), median progression-free survival (PFS), 6-months PFS and overall survival (OS). Treatment was safe with no dose-limiting toxicities or treatment-related deaths. The MTD has not been reached and, thus, 600 mg per week of NOX-A12 was established as RP2D for the ongoing expansion part of the trial. With increasing NOX-A12 dose levels, a corresponding increase of NOX-A12 plasma levels was observed. Of ten patients enrolled, nine showed radiographic responses, four reached partial remission. All but one patient (90%) showed at best response reduced perfusion values in terms of relative cerebral blood volume (rCBV). The median PFS was 174 (range 58-260) days, 6-month PFS was 40.0% and the median OS 389 (144-562) days. In a post-hoc exploratory analysis of tumor tissue, higher frequency of CXCL12+ endothelial and glioma cells was significantly associated with longer PFS under NOX-A12. Our data imply safety of NOX-A12 and its efficacy signal warrants further investigation.


Subject(s)
Aptamers, Nucleotide , Brain Neoplasms , Chemokine CXCL12 , Glioblastoma , Humans , Glioblastoma/radiotherapy , Glioblastoma/drug therapy , Aptamers, Nucleotide/administration & dosage , Chemokine CXCL12/blood , Male , Female , Middle Aged , Aged , Brain Neoplasms/radiotherapy , Brain Neoplasms/drug therapy , Adult , Maximum Tolerated Dose , Quality of Life , Neoplasm Recurrence, Local
3.
Cancer Med ; 13(10): e7286, 2024 May.
Article in English | MEDLINE | ID: mdl-38803199

ABSTRACT

OBJECTIVE: Radiotherapy (RT) is a cornerstone of the glioblastoma (GBM) treatment. However, the resistance of tumour cells to radiation results in early recurrence. The mechanisms underlying GBM radioresistance remain unclear. Screening for differentially expressed genes (DEGs) related to radiation might be a potential solution to this problem. METHOD: RT-associated DEGs were screened based on the RNA sequencing of 15 paired primary and recurrent GBMs. The mRNA and protein expression of candidate genes were validated in RNA sequencing of The Chinese Genome Atlas (CGGA) dataset and 18 cases of GBM samples. The relationship between the candidate gene and radiation was confirmed in irradiated GBM cells. The association of candidate gene with clinical characteristics and survival was investigated in the CGGA and TCGA dataset. Biological function and pathway analysis were explored by gene ontology analysis. The association of the candidate gene with radiosensitivity was verified using cell counting Kit-8, comet, and colony formation assays in vitro and subcutaneous tumour xenograft experiments in vivo. RESULTS: Gelsolin (GSN) was selected for further study. GSN expression was significant elevated in recurrent GBM and up-regulated in irradiated GBM cell lines. High expression of GSN was enriched in malignant phenotype of glioma. Moreover, high expression of GSN was associated with poor prognosis. Further investigation demonstrated that GSN-knockdown (GSN-KD) combined with RT significantly inhibited cell proliferation and enhanced radiosensitivity in vivo and in vitro. Mechanistically, GSN-KD could lead to more serious DNA damage and promotes apoptosis after RT. CONCLUSION: Radiation induced up-regulated of GSN. GSN-KD could enhance the radiosensitivity of GBM.


Subject(s)
Brain Neoplasms , Gelsolin , Gene Expression Regulation, Neoplastic , Glioblastoma , Radiation Tolerance , Humans , Glioblastoma/genetics , Glioblastoma/radiotherapy , Glioblastoma/pathology , Radiation Tolerance/genetics , Gelsolin/genetics , Animals , Mice , Cell Line, Tumor , Brain Neoplasms/genetics , Brain Neoplasms/radiotherapy , Brain Neoplasms/pathology , Gene Knockdown Techniques , Xenograft Model Antitumor Assays , Prognosis , Cell Proliferation , Apoptosis/genetics , Apoptosis/radiation effects , Male , Female , Mice, Nude , Neoplasm Recurrence, Local/genetics
4.
Sci Rep ; 14(1): 12363, 2024 05 29.
Article in English | MEDLINE | ID: mdl-38811596

ABSTRACT

Radiotherapy is the standard treatment for glioblastoma (GBM), but the overall survival rate for radiotherapy treated GBM patients is poor. The use of adjuvant and concomitant temozolomide (TMZ) improves the outcome; however, the effectiveness of this treatment varies according to MGMT levels. Herein, we evaluated whether MGMT expression affected the radioresponse of human GBM, GBM stem-like cells (GSCs), and melanoma. Our results indicated a correlation between MGMT promoter methylation status and MGMT expression. MGMT-producing cell lines ACPK1, GBMJ1, A375, and MM415 displayed enhanced radiosensitivity when MGMT was silenced using siRNA or when inhibited by lomeguatrib, whereas the OSU61, NSC11, WM852, and WM266-4 cell lines, which do not normally produce MGMT, displayed reduced radiosensitivity when MGMT was overexpressed. Mechanistically lomeguatrib prolonged radiation-induced γH2AX retention in MGMT-producing cells without specific cell cycle changes, suggesting that lomeguatrib-induced radiosensitization in these cells is due to radiation-induced DNA double-stranded break (DSB) repair inhibition. The DNA-DSB repair inhibition resulted in cell death via mitotic catastrophe in MGMT-producing cells. Overall, our results demonstrate that MGMT expression regulates radioresponse in GBM, GSC, and melanoma, implying a role for MGMT as a target for radiosensitization.


Subject(s)
DNA Modification Methylases , DNA Repair Enzymes , Glioblastoma , Melanoma , Radiation Tolerance , Tumor Suppressor Proteins , Humans , Glioblastoma/genetics , Glioblastoma/radiotherapy , Glioblastoma/metabolism , Glioblastoma/pathology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Melanoma/radiotherapy , DNA Modification Methylases/metabolism , DNA Modification Methylases/genetics , Cell Line, Tumor , Radiation Tolerance/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/radiation effects , Neoplastic Stem Cells/pathology , Promoter Regions, Genetic , DNA Methylation , DNA Repair , DNA Breaks, Double-Stranded/radiation effects , Gene Expression Regulation, Neoplastic , Temozolomide/pharmacology , Brain Neoplasms/genetics , Brain Neoplasms/radiotherapy , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Purines
5.
Int J Biol Macromol ; 270(Pt 1): 131782, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38734343

ABSTRACT

Glioblastoma is a rapidly fatal brain cancer that does not respond to therapy. Previous research showed that the transcriptional repressor protein BCL6 is upregulated by chemo and radiotherapy in glioblastoma, and inhibition of BCL6 enhances the effectiveness of these therapies. Therefore, BCL6 is a promising target to improve the efficacy of current glioblastoma treatment. BCL6 acts as a transcriptional repressor in germinal centre B cells and as an oncogene in lymphoma and other cancers. However, in glioblastoma, BCL6 induced by therapy may not be able to repress transcription. Using a BCL6 inhibitor, the whole proteome response to irradiation was compared with and without BCL6 activity. Acute high dose irradiation caused BCL6 to switch from repressing the DNA damage response to promoting stress response signalling. Rapid immunoprecipitation mass spectrometry of endogenous proteins (RIME) enabled comparison of BCL6 partner proteins between untreated and irradiated glioblastoma cells. BCL6 was associated with transcriptional coregulators in untreated glioblastoma including the known partner NCOR2. However, this association was lost in response to acute irradiation, where BCL6 unexpectedly associated with synaptic and plasma membrane proteins. These results reveal the activity of BCL6 under therapy-induced stress is context-dependent, and potentially altered by the intensity of that stress.


Subject(s)
Glioblastoma , Proto-Oncogene Proteins c-bcl-6 , Glioblastoma/radiotherapy , Glioblastoma/metabolism , Proto-Oncogene Proteins c-bcl-6/metabolism , Proto-Oncogene Proteins c-bcl-6/genetics , Humans , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/radiation effects , DNA Damage , Brain Neoplasms/radiotherapy , Brain Neoplasms/metabolism
6.
Sci Rep ; 14(1): 9283, 2024 04 23.
Article in English | MEDLINE | ID: mdl-38654028

ABSTRACT

We compared survival outcomes of high-dose concomitant boost radiotherapy (HDCBRT) and conventional dose radiotherapy (CRT) for newly diagnosed glioblastoma (GB). Patients treated with intensity-modulated radiation therapy for newly diagnosed GB were included. In HDCBRT, specific targets received 69, 60, and 51 Gy in 30 fractions, while 60 Gy in 30 fractions was administered with a standard radiotherapy method in CRT. Overall survival (OS) and progression-free survival (PFS) were compared using the Log-rank test, followed by multivariate Cox analysis. The inverse probability of treatment weighting (IPTW) method was also applied to each analysis. Among 102 eligible patients, 45 received HDCBRT and 57 received CRT. With a median follow-up of 16 months, the median survival times of OS and PFS were 21 and 9 months, respectively. No significant differences were observed in OS or PFS in the Kaplan-Meier analyses. In the multivariate analysis, HDCBRT correlated with improved OS (hazard ratio, 0.49; 95% confidence interval, 0.27-0.90; P = 0.021), and this result remained consistent after IPTW adjustments (P = 0.028). Conversely, dose suppression due to the proximity of normal tissues and IMRT field correlated with worse OS and PFS (P = 0.008 and 0.049, respectively). A prospective study with a stricter protocol is warranted to validate the efficacy of HDCBRT for GB.


Subject(s)
Brain Neoplasms , Glioblastoma , Radiotherapy, Intensity-Modulated , Humans , Glioblastoma/radiotherapy , Glioblastoma/mortality , Male , Female , Middle Aged , Aged , Radiotherapy, Intensity-Modulated/methods , Adult , Brain Neoplasms/radiotherapy , Brain Neoplasms/mortality , Radiotherapy Dosage , Kaplan-Meier Estimate , Progression-Free Survival , Treatment Outcome
7.
Int J Immunopathol Pharmacol ; 38: 3946320241249395, 2024.
Article in English | MEDLINE | ID: mdl-38687369

ABSTRACT

Background: Glioblastoma, a highly aggressive brain tumor, poses a significant clinical challenge, particularly in the context of radiotherapy. In this study, we aimed to explore infiltrating immune cells and identify immune-related genes associated with glioblastoma radiotherapy prognosis. Subsequently, we constructed a signature based on these genes to discern differences in molecular and tumor microenvironment immune characteristics, ultimately informing potential therapeutic strategies for patients with varying risk profiles. Methods: We leveraged UCSC Xena and CGGA gene expression profiles from post-radiotherapy glioblastoma as verification cohorts. Infiltration ratios were stratified into high and low groups based on the median value. Differential gene expression was determined through Limma differential analysis. A signature comprising four genes was constructed, guided by Gene Ontology (GO) functional enrichment results and Kaplan-Meier survival analysis. We evaluated differences in cell infiltration levels, Immune Score, Stromal Score, and ESTIMATE Score and their Pearson correlations with the signature. Spearman's correlation was computed between the signature and patient drug sensitivity (IC50), predicted using Genomics of Drug Sensitivity in Cancer (GDSC) and CCLE databases. Results: Notably, the infiltration of central memory CD8+T cells exhibited a significant correlation with glioblastoma radiotherapy prognosis. Samples were dichotomized into high- and low-risk groups based on the optimal signature threshold (2.466642). Kaplan-Meier (K-M) survival analysis revealed that the high-risk group experienced a significantly poorer prognosis (p = .0068), with AUC values exceeding 0.82 at 1, 3, and 5 years, underscoring the robust predictive potential of the signature scoring system. Independent validation sets substantiated the validity of the signature. Statistically significant differences in tumor microenvironments (p < .05) were observed between high- and low-risk groups, and these differences were significantly correlated with the signature (p < .05). Furthermore, there were significant correlations between high and low-risk groups regarding immune checkpoint expressions, Immune Prognostic Score (IPS), and Tumor Immune Dysfunction and Exclusion (TIDE) scores. Conclusion: The immune cell signature, comprising SDC-1, PLAUR, FN1, and CXCL13, holds promise as a predictive tool for assessing glioblastoma prognosis following radiotherapy. This signature also offers valuable guidance for tailoring treatment strategies, emphasizing its potential clinical relevance in improving patient outcomes.


Subject(s)
Brain Neoplasms , Glioblastoma , Tumor Microenvironment , Humans , Glioblastoma/genetics , Glioblastoma/immunology , Glioblastoma/radiotherapy , Glioblastoma/therapy , Glioblastoma/pathology , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/radiotherapy , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Prognosis , Tumor Microenvironment/immunology , Gene Expression Regulation, Neoplastic , Biomarkers, Tumor/genetics , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating/immunology , Gene Expression Profiling , Transcriptome , CD8-Positive T-Lymphocytes/immunology , Male
8.
J Neurooncol ; 168(1): 91-97, 2024 May.
Article in English | MEDLINE | ID: mdl-38598087

ABSTRACT

PURPOSE: Boron neutron capture therapy (BNCT) is a tumor cell-selective particle-radiation therapy. In BNCT, administered p-boronophenylalanine (BPA) is selectively taken up by tumor cells, and the tumor is irradiated with thermal neutrons. High-LET α-particles and recoil 7Li, which have a path length of 5-9 µm, are generated by the capture reaction between 10B and thermal neutrons and selectively kill tumor cells that have uptaken 10B. Although BNCT has prolonged the survival time of malignant glioma patients, recurrences are still to be resolved. miRNAs, that are encapsulated in small extracellular vesicles (sEVs) in body fluids and exist stably may serve critical role in recurrence. In this study, we comprehensively investigated microRNAs (miRNAs) in sEVs released from post-BNCT glioblastoma cells. METHOD: Glioblastoma U87 MG cells were treated with 25 ppm of BPA in the culture media and irradiated with thermal neutrons. After irradiation, they were plated into dishes and cultured for 3 days in the 5% CO2 incubator. Then, sEVs released into the medium were collected by column chromatography, and miRNAs in sEVs were comprehensively investigated using microarrays. RESULT: An increase in 20 individual miRNAs (ratio > 2) and a decrease in 2 individual miRNAs (ratio < 0.5) were detected in BNCT cells compared with non-irradiated cells. Among detected miRNAs, 20 miRNAs were associated with worse prognosis of glioma in Kaplan Meier Survival Analysis of overall survival in TCGA. CONCLUSION: These miRNA after BNCT may proceed tumors, modulate radiation resistance, or inhibit invasion and affect the prognosis of glioma.


Subject(s)
Boron Neutron Capture Therapy , Brain Neoplasms , Extracellular Vesicles , Glioblastoma , MicroRNAs , Boron Neutron Capture Therapy/methods , Humans , Extracellular Vesicles/metabolism , Extracellular Vesicles/radiation effects , MicroRNAs/metabolism , MicroRNAs/genetics , Glioblastoma/radiotherapy , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/genetics , Brain Neoplasms/radiotherapy , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/radiation effects
9.
Radiat Environ Biophys ; 63(2): 215-262, 2024 May.
Article in English | MEDLINE | ID: mdl-38664268

ABSTRACT

In the present research, we have developed a model-based crisp logic function statistical classifier decision support system supplemented with treatment planning systems for radiation oncologists in the treatment of glioblastoma multiforme (GBM). This system is based on Monte Carlo radiation transport simulation and it recreates visualization of treatment environments on mathematical anthropomorphic brain (MAB) phantoms. Energy deposition within tumour tissue and normal tissues are graded by quality audit factors which ensure planned dose delivery to tumour site thereby minimising damages to healthy tissues. The proposed novel methodology predicts tumour growth response to radiation therapy from a patient-specific medicine quality audit perspective. Validation of the study was achieved by recreating thirty-eight patient-specific mathematical anthropomorphic brain phantoms of treatment environments by taking into consideration density variation and composition of brain tissues. Dose computations accomplished through water phantom, tissue-equivalent head phantoms are neither cost-effective, nor patient-specific customized and is often less accurate. The above-highlighted drawbacks can be overcome by using open-source Electron Gamma Shower (EGSnrc) software and clinical case reports for MAB phantom synthesis which would result in accurate dosimetry with due consideration to the time factors. Considerable dose deviations occur at the tumour site for environments with intraventricular glioblastoma, haematoma, abscess, trapped air and cranial flaps leading to quality factors with a lower logic value of 0. Logic value of 1 depicts higher dose deposition within healthy tissues and also leptomeninges for majority of the environments which results in radiation-induced laceration.


Subject(s)
Brain Neoplasms , Glioblastoma , Monte Carlo Method , Glioblastoma/radiotherapy , Humans , Brain Neoplasms/radiotherapy , Phantoms, Imaging , Radiotherapy Planning, Computer-Assisted/methods , Radiation Oncologists , Decision Support Systems, Clinical , Radiotherapy Dosage
10.
Endokrynol Pol ; 75(2): 130-139, 2024.
Article in English | MEDLINE | ID: mdl-38646982

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive malignant brain tumour. The average survival time for a patient diagnosed with GBM, using standard treatment methods, is several months. Authors of the article pose a direct question: Is it possible to treat GBM solely with radioactive iodine (¹³¹I) therapy without employing the sodium iodide symporter (NIS) gene? After all, NIS has been detected not only in the thyroid but also in various tumours. The main author of this article (A.C.), with the assistance of her colleagues (physicians and pharmacologists), underwent ¹³¹I therapy after prior iodine inhibition, resulting in approximately 30% reduction in tumour size as revealed by magnetic resonance imaging (MRI). Classical therapy for GBM encompasses neurosurgery, conventional radiotherapy, and chemotherapy (e.g. temozolomide). Currently, tyrosine kinase inhibitors (imatinib, sunitinib, and sorafenib) are being used. Additionally, novel drugs such as crizotinib, entrectinib, or larotrectinib are being applied. Recently, personalised multimodal immunotherapy (IMI) based on anti-tumour vaccines derived from oncolytic viruses has been developed, concomitant with the advancement of cellular and molecular immunology. Thus, ¹³¹I therapy has been successfully employed for the first time in the case of GBM recurrence.


Subject(s)
Brain Neoplasms , Glioblastoma , Iodine Radioisotopes , Humans , Glioblastoma/radiotherapy , Glioblastoma/therapy , Glioblastoma/drug therapy , Iodine Radioisotopes/therapeutic use , Brain Neoplasms/radiotherapy , Brain Neoplasms/drug therapy , Brain Neoplasms/therapy , Neoplasm Recurrence, Local/prevention & control , Combined Modality Therapy
11.
Nat Commun ; 15(1): 3226, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38622132

ABSTRACT

The tumor microenvironment plays a crucial role in determining response to treatment. This involves a series of interconnected changes in the cellular landscape, spatial organization, and extracellular matrix composition. However, assessing these alterations simultaneously is challenging from a spatial perspective, due to the limitations of current high-dimensional imaging techniques and the extent of intratumoral heterogeneity over large lesion areas. In this study, we introduce a spatial proteomic workflow termed Hyperplexed Immunofluorescence Imaging (HIFI) that overcomes these limitations. HIFI allows for the simultaneous analysis of > 45 markers in fragile tissue sections at high magnification, using a cost-effective high-throughput workflow. We integrate HIFI with machine learning feature detection, graph-based network analysis, and cluster-based neighborhood analysis to analyze the microenvironment response to radiation therapy in a preclinical model of glioblastoma, and compare this response to a mouse model of breast-to-brain metastasis. Here we show that glioblastomas undergo extensive spatial reorganization of immune cell populations and structural architecture in response to treatment, while brain metastases show no comparable reorganization. Our integrated spatial analyses reveal highly divergent responses to radiation therapy between brain tumor models, despite equivalent radiotherapy benefit.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Mice , Proteomics , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/radiotherapy , Brain Neoplasms/pathology , Glioblastoma/diagnostic imaging , Glioblastoma/radiotherapy , Glioblastoma/pathology , Brain/pathology , Fluorescent Antibody Technique , Tumor Microenvironment
12.
Methods Cell Biol ; 185: 99-113, 2024.
Article in English | MEDLINE | ID: mdl-38556454

ABSTRACT

Radiotherapy is a crucial treatment modality for cancer patients, with approximately 60% of individuals undergoing ionizing radiation as part of their disease management. In recent years, there has been a growing trend toward minimizing irradiation fields through the use of image-guided dosimetry and innovative technologies. These advancements allow for selective irradiation, delivering higher local doses while reducing the number of treatment sessions. Consequently, computer-assisted methods have significantly enhanced the effectiveness of radiotherapy in the curative and palliative treatment of various cancers. Although radiation therapy alone can effectively achieve local control in some cancer types, it may not be sufficient for others. As a result, further preclinical research is necessary to explore novel approaches including new schedules of radiotherapy treatments. Unfortunately, there is a concerning lack of correlation between clinical outcomes and experiments conducted on mouse models. We hypothesize that this disparity arises from the differences in irradiation strategies employed in preclinical studies compared to those used in clinical practice, which ultimately affects the translatability of findings to patients. In this study, we present two comprehensive radiotherapy protocols for the treatment of orthotopic melanoma and glioblastoma tumors. These protocols utilize a small animal radiation research platform, which is an ideal radiation device for delivering localized and precise X-ray doses to the tumor mass. By employing these platforms, we aim to limit the side effects associated with irradiating healthy surrounding tissues. Our detailed protocols offer a valuable framework for conducting preclinical studies that closely mimic clinical radiotherapy techniques, bridging the gap between experimental results and patient outcomes.


Subject(s)
Glioblastoma , Radiotherapy, Image-Guided , Mice , Humans , Animals , Radiotherapy Planning, Computer-Assisted/methods , Radiotherapy, Image-Guided/methods , Glioblastoma/pathology , Glioblastoma/radiotherapy , Disease Models, Animal
13.
Neuro Oncol ; 26(12 Suppl 2): S3-S16, 2024 03 04.
Article in English | MEDLINE | ID: mdl-38437669

ABSTRACT

Chemoradiotherapy is the standard treatment after maximal safe resection for glioblastoma (GBM). Despite advances in molecular profiling, surgical techniques, and neuro-imaging, there have been no major breakthroughs in radiotherapy (RT) volumes in decades. Although the majority of recurrences occur within the original gross tumor volume (GTV), treatment of a clinical target volume (CTV) ranging from 1.5 to 3.0 cm beyond the GTV remains the standard of care. Over the past 15 years, the incorporation of standard and functional MRI sequences into the treatment workflow has become a routine practice with increasing adoption of MR simulators, and new integrated MR-Linac technologies allowing for daily pre-, intra- and post-treatment MR imaging. There is now unprecedented ability to understand the tumor dynamics and biology of GBM during RT, and safe CTV margin reduction is being investigated with the goal of improving the therapeutic ratio. The purpose of this review is to discuss margin strategies and the potential for adaptive RT for GBM, with a focus on the challenges and opportunities associated with both online and offline adaptive workflows. Lastly, opportunities to biologically guide adaptive RT using non-invasive imaging biomarkers and the potential to define appropriate volumes for dose modification will be discussed.


Subject(s)
Glioblastoma , Neurology , Radiation Oncology , Humans , Glioblastoma/radiotherapy , Chemoradiotherapy
14.
Neuro Oncol ; 26(12 Suppl 2): S17-S25, 2024 03 04.
Article in English | MEDLINE | ID: mdl-38437666

ABSTRACT

Advances in diagnostic and treatment technology along with rapid developments in translational research may now allow the realization of precision radiotherapy. Integration of biologically informed multimodality imaging to address the spatial and temporal heterogeneity underlying treatment resistance in glioblastoma is now possible for patient care, with evidence of safety and potential benefit. Beyond their diagnostic utility, several candidate imaging biomarkers have emerged in recent early-phase clinical trials of biologically based radiotherapy, and their definitive assessment in multicenter prospective trials is already in development. In this review, the rationale for clinical implementation of candidate advanced magnetic resonance imaging and positron emission tomography imaging biomarkers to guide personalized radiotherapy, the current landscape, and future directions for integrating imaging biomarkers into radiotherapy for glioblastoma are summarized. Moving forward, response-adaptive radiotherapy using biologically informed imaging biomarkers to address emerging treatment resistance in rational combination with novel systemic therapies may ultimately permit improvements in glioblastoma outcomes and true individualization of patient care.


Subject(s)
Glioblastoma , Radiation Oncology , Humans , Glioblastoma/diagnostic imaging , Glioblastoma/radiotherapy , Prospective Studies , Multimodal Imaging , Biomarkers , Multicenter Studies as Topic
15.
J Neurooncol ; 168(1): 49-56, 2024 May.
Article in English | MEDLINE | ID: mdl-38520571

ABSTRACT

BACKGROUND: The optimal management strategy for recurrent glioblastoma (rGBM) remains uncertain, and the impact of re-irradiation (Re-RT) on overall survival (OS) is still a matter of debate. This study included patients who achieved gross total resection (GTR) after a second surgery after recurrence, following the GlioCave criteria. METHODS: Inclusion criteria include being 18 years or older, having histologically confirmed locally recurrent IDHwt or IDH unknown GBM, achieving MRI-proven GTR after the second surgery, having a Karnofsky performance status of at least 60% after the second surgery, having a minimum interval of 6 months between the first radiotherapy and the second surgery, and a maximum of 8 weeks from second surgery to the start of Re-RT. RESULTS: A total of 44 patients have met the inclusion criteria. The median OS after the second surgery was 14 months. All patients underwent standard treatment after initial diagnosis, including maximum safe resection, adjuvant radiochemotherapy and adjuvant chemotherapy. Re-RT did not significantly impact OS. However, MGMT promoter methylation status and a longer interval (> 12 months) between treatments were associated with better OS. Multivariate analysis revealed the MGMT status as the only significant predictor of OS. CONCLUSION: Factors such as MGMT promoter methylation status and treatment interval play crucial roles in determining patient outcomes after second surgery. Personalized treatment strategies should consider these factors to optimize the management of rGBM. Prospective research is needed to define the value of re-RT after second surgery and to inform decision making in this situation.


Subject(s)
Brain Neoplasms , Glioblastoma , Neoplasm Recurrence, Local , Re-Irradiation , Humans , Glioblastoma/radiotherapy , Glioblastoma/surgery , Glioblastoma/mortality , Brain Neoplasms/radiotherapy , Brain Neoplasms/surgery , Brain Neoplasms/mortality , Male , Female , Middle Aged , Neoplasm Recurrence, Local/pathology , Aged , Adult , Re-Irradiation/methods , Cohort Studies , Radiotherapy, Adjuvant , Tertiary Care Centers , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
16.
BMJ Open ; 14(3): e078926, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38458809

ABSTRACT

INTRODUCTION: Glioblastoma (GBM) is the most common adult primary malignant brain tumour. The condition is incurable and, despite aggressive treatment at first presentation, almost all tumours recur after a median of 7 months. The aim of treatment at recurrence is to prolong survival and maintain health-related quality of life (HRQoL). Chemotherapy is typically employed for recurrent GBM, often using nitrosourea-based regimens. However, efficacy is limited, with reported median survivals between 5 and 9 months from recurrence. Although less commonly used in the UK, there is growing evidence that re-irradiation may produce survival outcomes at least similar to nitrosourea-based chemotherapy. However, there remains uncertainty as to the optimum approach and there is a paucity of available data, especially with regards to HRQoL. Brain Re-Irradiation Or Chemotherapy (BRIOChe) aims to assess re-irradiation, as an acceptable treatment option for recurrent IDH-wild-type GBM. METHODS AND ANALYSIS: BRIOChe is a phase II, multi-centre, open-label, randomised trial in patients with recurrent GBM. The trial uses Sargent's three-outcome design and will recruit approximately 55 participants from 10 to 15 UK radiotherapy sites, allocated (2:1) to receive re-irradiation (35 Gy in 10 daily fractions) or nitrosourea-based chemotherapy (up to six, 6-weekly cycles). The primary endpoint is overall survival rate for re-irradiation patients at 9 months. There will be no formal statistical comparison between treatment arms for the decision-making primary analysis. The chemotherapy arm will be used for calibration purposes, to collect concurrent data to aid interpretation of results. Secondary outcomes include HRQoL, dexamethasone requirement, anti-epileptic drug requirement, radiological response, treatment compliance, acute and late toxicities, progression-free survival. ETHICS AND DISSEMINATION: BRIOChe obtained ethical approval from Office for Research Ethics Committees Northern Ireland (reference no. 20/NI/0070). Final trial results will be published in peer-reviewed journals and adhere to the ICMJE guidelines. TRIAL REGISTRATION NUMBER: ISRCTN60524.


Subject(s)
Glioblastoma , Re-Irradiation , Adult , Humans , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Quality of Life , Neoplasm Recurrence, Local/drug therapy , Brain , Randomized Controlled Trials as Topic , Multicenter Studies as Topic , Clinical Trials, Phase II as Topic
17.
J Neurooncol ; 168(1): 69-76, 2024 May.
Article in English | MEDLINE | ID: mdl-38551747

ABSTRACT

PURPOSE: There remains no standard of care for patients with recurrent and chemorefractory glioblastoma. Re-irradiation (reRT) provides an additional management option. However, published series predominantly focus on small reRT volumes utilizing stereotactic hypofractionated regimens. Concerns regarding toxicity have limited utilisation of reRT for larger recurrences, however this may be mitigated with use of bevacizumab (BEV). METHODS AND MATERIALS: A prospective database of patients managed with the EORTC-NCIC (Stupp) protocol 60 Gy chemoradiotherapy protocol for glioblastoma between 2007 and 2021 was reviewed for those patients receiving reRT for chemorefractory relapse. Serial MRI and PET were used to establish true progression and exclude patients with pseudoprogression or radionecrosis from reRT. The primary endpoint was overall survival (OS) from date of reRT. Prognostic factors were also assessed. RESULTS: 447 patients managed for glioblastoma under the Stupp protocol were identified, of which 372 had relapsed and were thus eligible for reRT. 71 patients underwent reRT. Median relapse-free survival from diagnosis for the reRT and overall cohorts were similar at 11.6 months (95%CI:9.4-14.2) and 11.8 months (95%CI:9.4-14.2) respectively. 60/71 (85%) reRT patients had received BEV prior to reRT and continued concurrent BEV during reRT. Of the 11 patients not managed with BEV during reRT, 10 required subsequent salvage BEV. ReRT patients were younger (median 53 vs. 59 years, p < 0.001), had better performance status (86% vs. 69% ECOG 0-1, p = 0.002) and more commonly had MGMT promoter-methylated tumours (54% vs. 40%, p = 0.083) compared to non-reRT patients. Median reRT PTV volume was 135cm3 (IQR: 69-207cm3). Median OS from reRT to death was 7.1 months (95%CI:6.3-7.9). Patients aged < 50, 50-70 and > 70 years had post-reRT median OS of 7.7, 6.4 and 6.0 months respectively (p = 0.021). Median post-reRT survival was longer for patients with ECOG performance status 0-1 compared to 2-3 (8.1 vs. 6.3 months, p = 0.039). PTV volume, site of relapse, MGMT promoter-methylation status and extent of initial surgical resection were not associated with post-reRT survival. ReRT was well-tolerated. Out of the 6 patients (8%) admitted to hospital after reRT, only one was for reRT toxicity. This was a CTCAE grade 3 radiation necrosis event in a patient managed without prior BEV. CONCLUSION: Patients with recurrent glioblastoma who have been previously treated with 60 Gy radiotherapy have a meaningful survival benefit from large volume re-irradiation which is well tolerated. ReRT should not be ignored as a salvage treatment option in patients with chemorefractory progressive disease.


Subject(s)
Antineoplastic Agents, Immunological , Bevacizumab , Brain Neoplasms , Glioblastoma , Neoplasm Recurrence, Local , Radiation Dose Hypofractionation , Re-Irradiation , Humans , Glioblastoma/radiotherapy , Glioblastoma/drug therapy , Glioblastoma/therapy , Glioblastoma/pathology , Bevacizumab/therapeutic use , Bevacizumab/administration & dosage , Female , Male , Middle Aged , Brain Neoplasms/radiotherapy , Brain Neoplasms/drug therapy , Brain Neoplasms/therapy , Neoplasm Recurrence, Local/radiotherapy , Neoplasm Recurrence, Local/drug therapy , Re-Irradiation/methods , Aged , Antineoplastic Agents, Immunological/therapeutic use , Adult , Prospective Studies , Salvage Therapy , Retrospective Studies , Prognosis , Chemoradiotherapy/methods , Follow-Up Studies , Survival Rate
18.
Cell Prolif ; 57(6): e13606, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38454614

ABSTRACT

Glioblastoma (GBM), a WHO grade IV glioma, is a malignant primary brain tumour for which combination of surgery, chemotherapy and radiotherapy is the first-line approach despite adverse effects. Tumour microenvironment (TME) is characterized by an interplay of cells and soluble factors holding a critical role in neoplastic development. Significant pathophysiological changes have been found in GBM TME, such as glia activation and oxidative stress. Microglia play a crucial role in favouring GBM growth, representing target cells of immune escape mechanisms. Our study aims at analysing radiation-induced effects in modulating intercellular communication and identifying the basis of protective mechanisms in radiation-naïve GBM cells. Tumour cells were treated with conditioned media (CM) derived from 0, 2 or 15 Gy irradiated GBM cells or 0, 2 or 15 Gy irradiated human microglia. We demonstrated that irradiated microglia promote an increase of GBM cell lines proliferation through paracrine signalling. On the contrary, irradiated GBM-derived CM affect viability, triggering cell death mechanisms. In addition, we investigated whether these processes involve mitochondrial mass, fitness and oxidative phosphorylation and how GBM cells respond at these induced alterations. Our study suggests that off-target radiotherapy modulates microglia to support GBM proliferation and induce metabolic modifications.


Subject(s)
Brain Neoplasms , Cell Proliferation , Glioblastoma , Microglia , Tumor Microenvironment , Humans , Glioblastoma/radiotherapy , Glioblastoma/pathology , Glioblastoma/metabolism , Microglia/metabolism , Microglia/pathology , Microglia/radiation effects , Cell Proliferation/radiation effects , Cell Line, Tumor , Culture Media, Conditioned/pharmacology , Tumor Microenvironment/radiation effects , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Brain Neoplasms/metabolism , Cell Survival/radiation effects , Mitochondria/metabolism , Mitochondria/radiation effects
19.
Chin Clin Oncol ; 13(1): 11, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38453659

ABSTRACT

Glioblastoma is the most common primary brain tumor with an estimated 14,000 Americans diagnosed with this disease annually. This disease is treated with maximal surgical resection followed by adjuvant radiation therapy. Radiation therapy was initially delivered to the whole brain and with no concurrent or adjuvant systemic therapy. Advances in imaging and treatment delivery have allowed for partial brain irradiation to minimize radiation dose to normal structures, as well as sparing structures important for memory such as the hippocampus, decreasing morbidity and toxicity. While there is no consensus on the optimal radiation volume needed to successfully treat glioblastoma, there is consensus that the tumor bed with margin is preferable to treatment of the whole brain. Additionally, advances in knowledge regarding tumor biology have demonstrated the benefit of concurrent and adjuvant chemotherapy, as well as demonstrated that methylation of genes in the tumor can predispose greater responsiveness to chemotherapy. The following review describes the advancements in specific radiation techniques that have been used to improve the therapeutic ratio for management of glioblastoma and methods used to personalize radiation treatment for patients based on genomic markers as well as clinical factors. The review also describes future investigations that are currently taking place in order to enable a further improvement of clinical outcomes for patients with glioblastoma.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/genetics , Glioblastoma/radiotherapy , Glioblastoma/pathology , Combined Modality Therapy , Brain Neoplasms/genetics , Brain Neoplasms/radiotherapy , Brain Neoplasms/pathology , Chemotherapy, Adjuvant
SELECTION OF CITATIONS
SEARCH DETAIL
...