Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 34.880
Filtrer
1.
Cancer Immunol Immunother ; 73(9): 173, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38953982

RÉSUMÉ

Recent studies have indicated that combining oncolytic viruses with CAR-T cells in therapy has shown superior anti-tumor effects, representing a promising approach. Nonetheless, the localized delivery method of intratumoral injection poses challenges for treating metastatic tumors or distal tumors that are difficult to reach. To address this obstacle, we employed HSV-1-infected CAR-T cells, which systemically delivery HSV into solid tumors. The biological function of CAR-T cells remained intact after loading them with HSV for a period of three days. In both immunocompromised and immunocompetent GBM orthotopic mouse models, B7-H3 CAR-T cells effectively delivered HSV to tumor lesions, resulting in enhanced T-cell infiltration and significantly prolonged survival in mice. We also employed a bilateral subcutaneous tumor model and observed that the group receiving intratumoral virus injection exhibited a significant reduction in tumor volume on the injected side, while the group receiving intravenous infusion of CAR-T cells carrying HSV displayed suppressed tumor growth on both sides. Hence, CAR-THSV cells offer notable advantages in the systemic delivery of HSV to distant tumors. In conclusion, our findings emphasize the potential of CAR-T cells as carriers for HSV, presenting significant advantages for oncolytic virotherapy targeting distant tumors.


Sujet(s)
Immunothérapie adoptive , Thérapie virale de cancers , Virus oncolytiques , Récepteurs chimériques pour l'antigène , Animaux , Souris , Thérapie virale de cancers/méthodes , Humains , Virus oncolytiques/immunologie , Virus oncolytiques/génétique , Immunothérapie adoptive/méthodes , Récepteurs chimériques pour l'antigène/immunologie , Herpèsvirus humain de type 1/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe , Lignée cellulaire tumorale , Lymphocytes T/immunologie , Femelle , Glioblastome/thérapie , Glioblastome/immunologie
2.
PLoS One ; 19(7): e0306142, 2024.
Article de Anglais | MEDLINE | ID: mdl-38954698

RÉSUMÉ

Developing T1-weighted magnetic resonance imaging (MRI) contrast agents with enhanced biocompatibility and targeting capabilities is crucial owing to concerns over current agents' potential toxicity and suboptimal performance. Drawing inspiration from "biomimetic camouflage," we isolated cell membranes (CMs) from human glioblastoma (T98G) cell lines via the extrusion method to facilitate homotypic glioma targeting. At an 8:1 mass ratio of ferric chloride hexahydrate to gallic acid (GA), the resulting iron (Fe)-GA nanoparticles (NPs) proved effective as a T1-weighted MRI contrast agent. T98G CM-coated Fe-GA NPs demonstrated improved homotypic glioma targeting, validated through Prussian blue staining and in vitro MRI. This biomimetic camouflage strategy holds promise for the development of targeted theranostic agents in a safe and effective manner.


Sujet(s)
Produits de contraste , Acide gallique , Imagerie par résonance magnétique , Acide gallique/composition chimique , Humains , Imagerie par résonance magnétique/méthodes , Lignée cellulaire tumorale , Produits de contraste/composition chimique , Fer/composition chimique , Matériaux biomimétiques/composition chimique , Glioblastome/traitement médicamenteux , Glioblastome/imagerie diagnostique , Glioblastome/anatomopathologie , Nanoparticules/composition chimique , Composés du fer III/composition chimique , Membrane cellulaire/métabolisme
3.
Sci Rep ; 14(1): 15065, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956384

RÉSUMÉ

This study aimed to apply pathomics to predict Matrix metalloproteinase 9 (MMP9) expression in glioblastoma (GBM) and investigate the underlying molecular mechanisms associated with pathomics. Here, we included 127 GBM patients, 78 of whom were randomly allocated to the training and test cohorts for pathomics modeling. The prognostic significance of MMP9 was assessed using Kaplan-Meier and Cox regression analyses. PyRadiomics was used to extract the features of H&E-stained whole slide images. Feature selection was performed using the maximum relevance and minimum redundancy (mRMR) and recursive feature elimination (RFE) algorithms. Prediction models were created using support vector machines (SVM) and logistic regression (LR). The performance was assessed using ROC analysis, calibration curve assessment, and decision curve analysis. MMP9 expression was elevated in patients with GBM. This was an independent prognostic factor for GBM. Six features were selected for the pathomics model. The area under the curves (AUCs) of the training and test subsets were 0.828 and 0.808, respectively, for the SVM model and 0.778 and 0.754, respectively, for the LR model. The C-index and calibration plots exhibited effective estimation abilities. The pathomics score calculated using the SVM model was highly correlated with overall survival time. These findings indicate that MMP9 plays a crucial role in GBM development and prognosis. Our pathomics model demonstrated high efficacy for predicting MMP9 expression levels and prognosis of patients with GBM.


Sujet(s)
Glioblastome , Apprentissage machine , Matrix metalloproteinase 9 , Humains , Glioblastome/anatomopathologie , Glioblastome/mortalité , Glioblastome/métabolisme , Matrix metalloproteinase 9/métabolisme , Mâle , Femelle , Adulte d'âge moyen , Pronostic , Sujet âgé , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/mortalité , Machine à vecteur de support , Adulte , Estimation de Kaplan-Meier , Courbe ROC , Marqueurs biologiques tumoraux/métabolisme
4.
Mol Brain ; 17(1): 42, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956588

RÉSUMÉ

Glioblastoma (GBM) is an aggressive nervous system tumor with a poor prognosis. Although, surgery, radiation therapy, and chemotherapy are the current standard protocol for GBM patients, there is still a poor prognosis in these patients. Temozolomide (TMZ) as a first-line therapeutic agent in GBM can easily cross from the blood-brain barrier to inhibit tumor cell proliferation. However, there is a high rate of TMZ resistance in GBM patients. Since, there are limited therapeutic choices for GBM patients who develop TMZ resistance; it is required to clarify the molecular mechanisms of chemo resistance to introduce the novel therapeutic targets. MicroRNAs (miRNAs) regulate chemo resistance through regulation of drug metabolism, absorption, DNA repair, apoptosis, and cell cycle. In the present review we discussed the role of miRNAs in TMZ response of GBM cells. It has been reported that miRNAs mainly induced TMZ sensitivity by regulation of signaling pathways and autophagy in GBM cells. Therefore, miRNAs can be used as the reliable diagnostic/prognostic markers in GBM patients. They can also be used as the therapeutic targets to improve the TMZ response in GBM cells.


Sujet(s)
Tumeurs du cerveau , Résistance aux médicaments antinéoplasiques , Glioblastome , microARN , Témozolomide , Humains , Témozolomide/pharmacologie , Témozolomide/usage thérapeutique , Glioblastome/génétique , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , microARN/génétique , microARN/métabolisme , Résistance aux médicaments antinéoplasiques/génétique , Tumeurs du cerveau/génétique , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Animaux , Dacarbazine/analogues et dérivés , Dacarbazine/usage thérapeutique , Dacarbazine/pharmacologie , Autophagie/effets des médicaments et des substances chimiques , Autophagie/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
5.
PeerJ ; 12: e17579, 2024.
Article de Anglais | MEDLINE | ID: mdl-38978755

RÉSUMÉ

Background: Lysyl oxidase enzymes (LOXs), as extracellular matrix (ECM) protein regulators, play vital roles in tumor progression by remodeling the tumor microenvironment. However, their roles in glioblastoma (GBM) have not been fully elucidated. Methods: The genetic alterations and prognostic value of LOXs were investigated via cBioPortal. The correlations between LOXs and biological functions/molecular tumor subtypes were explored in The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA). After Kaplan‒Meier and Cox survival analyses, a Loxl1-based nomogram and prognostic risk score model (PRSM) were constructed and evaluated by time-dependent receiver operating characteristic curves, calibration curves, and decision curve analyses. Tumor enrichment pathways and immune infiltrates were explored by single-cell RNA sequencing and TIMER. Loxl1-related changes in tumor viability/proliferation and invasion were further validated by CCK-8, western blot, wound healing, and Transwell invasion assays. Results: GBM patients with altered LOXs had poor survival. Upregulated LOXs were found in IDH1-wildtype and mesenchymal (not Loxl1) GBM subtypes, promoting ECM receptor interactions in GBM. The Loxl1-based nomogram and the PRSM showed high accuracy, reliability, and net clinical benefits. Loxl1 expression was related to tumor invasion and immune infiltration (B cells, neutrophils, and dendritic cells). Loxl1 knockdown suppressed GBM cell proliferation and invasion by inhibiting the EMT pathway (through the downregulation of N-cadherin/Vimentin/Snai1 and the upregulation of E-cadherin). Conclusion: The Loxl1-based nomogram and PRSM were stable and individualized for assessing GBM patient prognosis, and the invasive role of Loxl1 could provide a promising therapeutic strategy.


Sujet(s)
Tumeurs du cerveau , Transition épithélio-mésenchymateuse , Glioblastome , Invasion tumorale , Humains , Glioblastome/anatomopathologie , Glioblastome/génétique , Glioblastome/mortalité , Glioblastome/métabolisme , Transition épithélio-mésenchymateuse/génétique , Pronostic , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/mortalité , Tumeurs du cerveau/métabolisme , Lignée cellulaire tumorale , Nomogrammes , Récepteurs éboueurs de classe E/métabolisme , Récepteurs éboueurs de classe E/génétique , Mâle , Microenvironnement tumoral , Femelle , Amino-acid oxidoreductases/génétique , Amino-acid oxidoreductases/métabolisme , Prolifération cellulaire , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Régulation de l'expression des gènes tumoraux , Lysyloxidase/métabolisme , Lysyloxidase/génétique , Isocitrate dehydrogenases/génétique , Isocitrate dehydrogenases/métabolisme
6.
Cancer Cell ; 42(7): 1217-1238.e19, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38981438

RÉSUMÉ

Although genomic anomalies in glioblastoma (GBM) have been well studied for over a decade, its 5-year survival rate remains lower than 5%. We seek to expand the molecular landscape of high-grade glioma, composed of IDH-wildtype GBM and IDH-mutant grade 4 astrocytoma, by integrating proteomic, metabolomic, lipidomic, and post-translational modifications (PTMs) with genomic and transcriptomic measurements to uncover multi-scale regulatory interactions governing tumor development and evolution. Applying 14 proteogenomic and metabolomic platforms to 228 tumors (212 GBM and 16 grade 4 IDH-mutant astrocytoma), including 28 at recurrence, plus 18 normal brain samples and 14 brain metastases as comparators, reveals heterogeneous upstream alterations converging on common downstream events at the proteomic and metabolomic levels and changes in protein-protein interactions and glycosylation site occupancy at recurrence. Recurrent genetic alterations and phosphorylation events on PTPN11 map to important regulatory domains in three dimensions, suggesting a central role for PTPN11 signaling across high-grade gliomas.


Sujet(s)
Tumeurs du cerveau , Gliome , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Transduction du signal , Humains , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Gliome/génétique , Gliome/anatomopathologie , Gliome/métabolisme , Mutation , Protéomique/méthodes , Maturation post-traductionnelle des protéines , Régulation de l'expression des gènes tumoraux , Glioblastome/génétique , Glioblastome/anatomopathologie , Glioblastome/métabolisme , Phosphorylation , Grading des tumeurs , Isocitrate dehydrogenases/génétique , Isocitrate dehydrogenases/métabolisme
7.
Oncol Res ; 32(7): 1173-1184, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948026

RÉSUMÉ

Background: Inhibitor of NF-κB kinase-interacting protein (IKIP) is known to promote proliferation of glioblastoma (GBM) cells, but how it affects migration and invasion by those cells is unclear. Methods: We compared levels of IKIP between glioma tissues and normal brain tissue in clinical samples and public databases. We examined the effects of IKIP overexpression and knockdown on the migration and invasion of GBM using transwell and wound healing assays, and we compared the transcriptomes under these different conditions to identify the molecular mechanisms involved. Results: Based on data from our clinical samples and from public databases, IKIP was overexpressed in GBM tumors, and its expression level correlated inversely with survival. IKIP overexpression in GBM cells inhibited migration and invasion in transwell and wound healing assays, whereas IKIP knockdown exerted the opposite effects. IKIP overexpression in GBM cells that were injected into mouse brain promoted tumor growth but inhibited tumor invasion of surrounding tissue. The effects of IKIP were associated with downregulation of THBS1 mRNA and concomitant inhibition of THBS1/FAK signaling. Conclusions: IKIP inhibits THBS1/FAK signaling to suppress migration and invasion of GBM cells.


Sujet(s)
Tumeurs du cerveau , Mouvement cellulaire , Focal adhesion kinase 1 , Glioblastome , Invasion tumorale , Transduction du signal , Thrombospondine-1 , Humains , Glioblastome/anatomopathologie , Glioblastome/métabolisme , Glioblastome/génétique , Animaux , Souris , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/génétique , Lignée cellulaire tumorale , Thrombospondine-1/métabolisme , Thrombospondine-1/génétique , Focal adhesion kinase 1/métabolisme , Focal adhesion kinase 1/génétique , Régulation négative , Régulation de l'expression des gènes tumoraux , Prolifération cellulaire
8.
Commun Biol ; 7(1): 793, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38951146

RÉSUMÉ

Brown and brown-like adipose tissues have attracted significant attention for their role in metabolism and therapeutic potential in diabetes and obesity. Despite compelling evidence of an interplay between adipocytes and lymphocytes, the involvement of these tissues in immune responses remains largely unexplored. This study explicates a newfound connection between neuroinflammation and brown- and bone marrow adipose tissue. Leveraging the use of [18F]F-AraG, a mitochondrial metabolic tracer capable of tracking activated lymphocytes and adipocytes simultaneously, we demonstrate, in models of glioblastoma and multiple sclerosis, the correlation between intracerebral immune infiltration and changes in brown- and bone marrow adipose tissue. Significantly, we show initial evidence that a neuroinflammation-adipose tissue link may also exist in humans. This study proposes the concept of an intricate immuno-neuro-adipose circuit, and highlights brown- and bone marrow adipose tissue as an intermediary in the communication between the immune and nervous systems. Understanding the interconnectedness within this circuitry may lead to advancements in the treatment and management of various conditions, including cancer, neurodegenerative diseases and metabolic disorders.


Sujet(s)
Tissu adipeux brun , Maladies neuro-inflammatoires , Animaux , Humains , Tissu adipeux brun/métabolisme , Maladies neuro-inflammatoires/immunologie , Maladies neuro-inflammatoires/métabolisme , Maladies neuro-inflammatoires/anatomopathologie , Moelle osseuse/métabolisme , Souris , Mâle , Glioblastome/anatomopathologie , Glioblastome/immunologie , Glioblastome/métabolisme , Souris de lignée C57BL , Femelle , Sclérose en plaques/anatomopathologie , Sclérose en plaques/immunologie , Sclérose en plaques/métabolisme , Sclérose en plaques/imagerie diagnostique , Tomographie par émission de positons
9.
PLoS One ; 19(7): e0301739, 2024.
Article de Anglais | MEDLINE | ID: mdl-38968207

RÉSUMÉ

With current imaging, discriminating tumor progression from treatment effect following immunotherapy or oncolytic virotherapy of glioblastoma (GBM) is challenging. A blood based diagnostic biomarker would therefore be helpful. Axl is a receptor tyrosine kinase that is highly expressed by many cancers including GBM. Axl expression is regulated through enzymatic cleavage of its extracellular domain. The resulting fragment can be detected in serum as soluble Axl (sAxl). sAxl levels can distinguish patients with melanoma, hepatocellular carcinoma, and pancreatic ductal adenocarcinoma from healthy controls. This is a pilot study to determine if sAxl is a candidate biomarker for GBM. The sAxl levels in the serum of 40 healthy volunteers and 20 GBM patients were determined using an enzyme-linked immunosorbent assay (ELISA). Pre- and post- operative sAxl levels were obtained. Volumetric MRI evaluation provided GBM tumor volume metrics. There was no significant difference in the sAxl levels of the volunteers (30.16±1.88 ng/ml) and GBM patients (30.74±1.96 ng/ml) p = 0.27. The postoperative sAxl levels were significantly higher than preoperative levels (32.32±2.26 ng/ml vs 30.74±1.96 ng/ml, p = 0.03). We found no correlation between tumor volume and sAxl levels. Axl expression was low or absent in 6 of 11 (55%) patient derived GBM cell lines. Given the wide range of Axl expression by GBM tumors, sAxl may not be a reliable indicator of GBM. However, given the small sample size in this study, a larger study may be considered.


Sujet(s)
Axl Receptor Tyrosine Kinase , Marqueurs biologiques tumoraux , Glioblastome , Protéines proto-oncogènes , Récepteurs à activité tyrosine kinase , Humains , Récepteurs à activité tyrosine kinase/métabolisme , Glioblastome/métabolisme , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/sang , Projets pilotes , Marqueurs biologiques tumoraux/sang , Marqueurs biologiques tumoraux/métabolisme , Mâle , Femelle , Adulte d'âge moyen , Adulte , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/sang , Sujet âgé , Imagerie par résonance magnétique/méthodes , Études cas-témoins , Test ELISA
10.
Cancer Immunol Immunother ; 73(9): 178, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38954031

RÉSUMÉ

Intracranial tumors present a significant therapeutic challenge due to their physiological location. Immunotherapy presents an attractive method for targeting these intracranial tumors due to relatively low toxicity and tumor specificity. Here we show that SCIB1, a TRP-2 and gp100 directed ImmunoBody® DNA vaccine, generates a strong TRP-2 specific immune response, as demonstrated by the high number of TRP2-specific IFNγ spots produced and the detection of a significant number of pentamer positive T cells in the spleen of vaccinated mice. Furthermore, vaccine-induced T cells were able to recognize and kill B16HHDII/DR1 cells after a short in vitro culture. Having found that glioblastoma multiforme (GBM) expresses significant levels of PD-L1 and IDO1, with PD-L1 correlating with poorer survival in patients with the mesenchymal subtype of GBM, we decided to combine SCIB1 ImmunoBody® with PD-1 immune checkpoint blockade to treat mice harboring intracranial tumors expressing TRP-2 and gp100. Time-to-death was significantly prolonged, and this correlated with increased CD4+ and CD8+ T cell infiltration in the tissue microenvironment (TME). However, in addition to PD-L1 and IDO, the GBM TME was found to contain a significant number of immunoregulatory T (Treg) cell-associated transcripts, and the presence of such cells is likely to significantly affect clinical outcome unless also tackled.


Sujet(s)
Tumeurs du cerveau , Vaccins anticancéreux , Inhibiteurs de points de contrôle immunitaires , Récepteur-1 de mort cellulaire programmée , Vaccins à ADN , Animaux , Femelle , Humains , Souris , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/immunologie , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/thérapie , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Lignée cellulaire tumorale , Glioblastome/immunologie , Glioblastome/thérapie , Glioblastome/traitement médicamenteux , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Immunothérapie/méthodes , Intramolecular oxidoreductases , Souris de lignée C57BL , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Vaccins à ADN/immunologie , Vaccins à ADN/usage thérapeutique , Mâle , Enfant , Adulte d'âge moyen
11.
Cells ; 13(13)2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38994929

RÉSUMÉ

Standard-of-care treatment for Glioblastoma Multiforme (GBM) is comprised of surgery and adjuvant chemoradiation. Chimeric Antigen Receptor (CAR) T cell therapy has demonstrated disease-modifying activity in GBM and holds great promise. Radiation, a standard-of-care treatment for GBM, has well-known immunomodulatory properties and may overcome the immunosuppressive tumor microenvironment (TME); however, radiation dose optimization and integration with CAR T cell therapy is not well defined. Murine immunocompetent models of GBM were treated with titrated doses of stereotactic radiosurgery (SRS) of 5, 10, and 20 Gray (Gy), and the TME was analyzed using Nanostring. A conditioning dose of 10 Gy was determined based on tumor growth kinetics and gene expression changes in the TME. We demonstrate that a conditioning dose of 10 Gy activates innate and adaptive immune cells in the TME. Mice treated with 10 Gy in combination with mCAR T cells demonstrated enhanced antitumor activity and superior memory responses to rechallenge with IL13Rα2-positive tumors. Furthermore, 10 Gy plus mCAR T cells also protected against IL13Rα2-negative tumors through a mechanism that was, in part, c-GAS-STING pathway-dependent. Together, these findings support combination conditioning with low-dose 10 Gy radiation in combination with mCAR T cells as a therapeutic strategy for GBM.


Sujet(s)
Glioblastome , Récepteurs chimériques pour l'antigène , Microenvironnement tumoral , Glioblastome/thérapie , Glioblastome/immunologie , Glioblastome/radiothérapie , Glioblastome/anatomopathologie , Animaux , Récepteurs chimériques pour l'antigène/métabolisme , Récepteurs chimériques pour l'antigène/immunologie , Souris , Microenvironnement tumoral/immunologie , Humains , Lignée cellulaire tumorale , Immunothérapie adoptive/méthodes , Tumeurs du cerveau/thérapie , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/radiothérapie , Lymphocytes T/immunologie , Souris de lignée C57BL , Immunomodulation , Femelle
12.
Medicine (Baltimore) ; 103(28): e38913, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38996177

RÉSUMÉ

The objective of this study was to assess the prognostic relevance of Stanniocalcin-2 (STC2) expression, as determined via immunohistochemistry in tumor tissue, in a cohort of 83 patients diagnosed with glioblastoma who underwent maximal safe surgical resection followed by radiotherapy concurrent with adjuvant temozolomide. STC2 expression levels were categorized using a 3-tiered semiquantitative system: negative expression (level 0-), low expression (level 1+), and high expression (levels 2 + and 3+). Patients were categorized into 2 distinct groups according to their STC2 expression levels: negative STC2 (-/+) and positive STC2 (++/+++). The primary outcome measure was the relationship between STC2 expression and progression-free survival (PFS), with overall survival (OS) serving as the secondary endpoint. Kaplan-Meier survival analysis confirmed that patients exhibiting high STC2 expression had significantly shorter OS (8 vs 20 months, P < .001) and PFS (6 vs 18 months, P < .001) than those with low or negative STC2 expression. Multivariate analysis revealed that STC2 expression was an independent prognostic factor for both OS (hazard ratio: 0.4; 95% confidence interval: 0.2-0.8; P < .05) and PFS (hazard ratio: 0.3; 95% confidence interval: 0.2-0.4; P < .05) in patients with glioblastoma. Furthermore, elevated STC2 expression in GBM was correlated with several established aggressive clinicopathological characteristics, including advanced age (≥65 years), low ECOG PS (≥2), and isocitrate dehydrogenase mutation negativity. These findings underscore that heightened STC2 expression within the tumor tissue of GBM patients functions as an adverse prognostic marker, correlating with an elevated risk of progression and reduced OS. Therapeutic interventions targeting the AKT-mTOR, ERK1-2, and mitogen-activated protein kinase pathways as well as immune checkpoint inhibitors and vascular endothelial growth factor blockade, as well as potential forthcoming antibody-drug conjugates targeting the STC2 molecule, have the potential to broaden the scope of combined treatment strategies.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs du cerveau , Glioblastome , Glycoprotéines , Protéines et peptides de signalisation intercellulaire , Humains , Glioblastome/métabolisme , Glioblastome/mortalité , Glioblastome/génétique , Glioblastome/anatomopathologie , Femelle , Mâle , Adulte d'âge moyen , Marqueurs biologiques tumoraux/métabolisme , Glycoprotéines/métabolisme , Pronostic , Sujet âgé , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/mortalité , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Protéines et peptides de signalisation intercellulaire/métabolisme , Protéines et peptides de signalisation intercellulaire/génétique , Adulte , Témozolomide/usage thérapeutique , Estimation de Kaplan-Meier , Survie sans progression , Études rétrospectives , Antinéoplasiques alcoylants/usage thérapeutique , Immunohistochimie
13.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-39000485

RÉSUMÉ

Cytotoxic activity has been reported for the xanthone α-mangostin (AMN) against Glioblastoma multiforme (GBM), an aggressive malignant brain cancer with a poor prognosis. Recognizing that AMN's high degree of hydrophobicity is likely to limit its systemic administration, we formulated AMN using reconstituted high-density lipoprotein (rHDL) nanoparticles. The photophysical characteristics of the formulation, including fluorescence lifetime and steady-state anisotropy, indicated that AMN was successfully incorporated into the rHDL nanoparticles. To our knowledge, this is the first report on the fluorescent characteristics of AMN with an HDL-based drug carrier. Cytotoxicity studies in a 2D culture and 3D spheroid model of LN-229 GBM cells and normal human astrocytes showed an enhanced therapeutic index with the rHDL-AMN formulation compared to the unincorporated AMN and Temozolomide, a standard GBM chemotherapy agent. Furthermore, treatment with the rHDL-AMN facilitated a dose-dependent upregulation of autophagy and reactive oxygen species generation to a greater extent in LN-229 cells compared to astrocytes, indicating the reduced off-target toxicity of this novel formulation. These studies indicate the potential therapeutic benefits to GBM patients via selective targeting using the rHDL-AMN formulation.


Sujet(s)
Glioblastome , Lipoprotéines HDL , Nanoparticules , Sphéroïdes de cellules , Xanthones , Humains , Xanthones/composition chimique , Xanthones/pharmacologie , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Glioblastome/métabolisme , Lignée cellulaire tumorale , Nanoparticules/composition chimique , Lipoprotéines HDL/composition chimique , Lipoprotéines HDL/métabolisme , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Vecteurs de médicaments/composition chimique , Espèces réactives de l'oxygène/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Astrocytes/métabolisme , Astrocytes/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Autophagie/effets des médicaments et des substances chimiques
14.
Cell Death Dis ; 15(7): 503, 2024 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-39003252

RÉSUMÉ

Glioblastoma multiforme (GBM) is the most common adult primary brain tumor. The standard clinical treatment of GBM includes a maximal surgical resection followed by concomitant radiotherapy (RT) and chemotherapy sessions with Temozolomide (TMZ) in addition to adjuvant TMZ cycles. Despite the severity of this protocol, GBM is highly resistant and recurs in almost all cases while the protocol remains unchanged since 2005. Limited-diffusion or chronic hypoxia has been identified as one of the major key players driving this aggressive phenotype. The presence of hypoxia within the tumor bulk contributes to the activation of hypoxia signaling pathway mediated by the hypoxia-inducing factors (HIFs), which in turn activate biological mechanisms to ensure the adaptation and survival of GBM under limited oxygen and nutrient supply. Activated downstream pathways are involved in maintaining stem cell-like phenotype, inducing mesenchymal shift, invasion, and migration, altering the cellular and oxygen metabolism, and increasing angiogenesis, autophagy, and immunosuppression. Therefore, in this review will discuss the recent preclinical and clinical approaches that aim at targeting tumor hypoxia to enhance the response of GBM to conventional therapies along with their results and limitations upon clinical translation.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Humains , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/thérapie , Animaux , Hypoxie cellulaire , Hypoxie tumorale , Transduction du signal
15.
Front Immunol ; 15: 1420182, 2024.
Article de Anglais | MEDLINE | ID: mdl-39007144

RÉSUMÉ

Introduction: Glioblastoma multiforme (GBM), the most common primary malignant brain tumor, is notorious for its aggressive growth and dismal prognosis. This study aimed to elucidate the molecular underpinnings of GBM, particularly focusing on the role of AGBL4 and its connection to inflammatory pathways, to discover viable therapeutic targets. Methods: Single-cell sequencing was utilized to examine the expression levels of AGBL4 and functional assays were performed to assess the effects of AGBL4 modulation. Results: Our findings identified the significant upregulation of AGBL4 in GBM, which correlated with adverse clinical outcomes. Functional assays demonstrated that AGBL4 knockdown inhibited GBM cell proliferation, migration, and invasion and influenced inflammatory response pathways, while AGBL4 overexpression promoted these activities. Further investigation revealed that AGBL4 exerted its oncogenic effects through modulation of MMP-1, establishing a novel regulatory axis critical for GBM progression and inflammation. Discussion: Both AGBL4 and MMP-1 may be pivotal molecular targets, offering new avenues for targeted therapy in GBM management.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Matrix metalloproteinase 1 , Glioblastome/anatomopathologie , Glioblastome/métabolisme , Glioblastome/génétique , Humains , Matrix metalloproteinase 1/métabolisme , Matrix metalloproteinase 1/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/génétique , Tumeurs du cerveau/immunologie , Lignée cellulaire tumorale , Prolifération cellulaire , Mouvement cellulaire/génétique , Évolution de la maladie , Inflammation/métabolisme , Régulation de l'expression des gènes tumoraux , Transduction du signal , Mâle
16.
ACS Nano ; 18(28): 18712-18728, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38952208

RÉSUMÉ

Immunotherapy can potentially suppress the highly aggressive glioblastoma (GBM) by promoting T lymphocyte infiltration. Nevertheless, the immune privilege phenomenon, coupled with the generally low immunogenicity of vaccines, frequently hampers the presence of lymphocytes within brain tumors, particularly in brain tumors. In this study, the membrane-disrupted polymer-wrapped CuS nanoflakes that can penetrate delivery to deep brain tumors via releasing the cell-cell interactions, facilitating the near-infrared II (NIR II) photothermal therapy, and detaining dendritic cells for a self-cascading immunotherapy are developed. By convection-enhanced delivery, membrane-disrupted amphiphilic polymer micelles (poly(methoxypoly(ethylene glycol)-benzoic imine-octadecane, mPEG-b-C18) with CuS nanoflakes enhances tumor permeability and resides in deep brain tumors. Under low-power NIR II irradiation (0.8 W/cm2), the intense heat generated by well-distributed CuS nanoflakes actuates the thermolytic efficacy, facilitating cell apoptosis and the subsequent antigen release. Then, the positively charged polymer after hydrolysis of the benzoic-imine bond serves as an antigen depot, detaining autologous tumor-associated antigens and presenting them to dendritic cells, ensuring sustained immune stimulation. This self-cascading penetrative immunotherapy amplifies the immune response to postoperative brain tumors but also enhances survival outcomes through effective brain immunotherapy.


Sujet(s)
Tumeurs du cerveau , Membrane cellulaire , Cellules dendritiques , Immunothérapie , Rayons infrarouges , Cellules dendritiques/immunologie , Cellules dendritiques/effets des médicaments et des substances chimiques , Tumeurs du cerveau/thérapie , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/anatomopathologie , Animaux , Souris , Humains , Membrane cellulaire/composition chimique , Lignée cellulaire tumorale , Micelles , Nanoparticules/composition chimique , Thérapie photothermique , Polyéthylène glycols/composition chimique , Glioblastome/thérapie , Glioblastome/immunologie , Glioblastome/anatomopathologie , Apoptose/effets des médicaments et des substances chimiques
17.
Bull Exp Biol Med ; 177(1): 147-154, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38963598

RÉSUMÉ

Hyaluronidase increases tissue permeability and diffusion of the extracellular fluid by cleaving hyaluronan, the primary component of the extracellular matrix. Hyaluronidase pegylation (Hyal-PEG) decreases its clearance and enhances biodistribution. The pro- and anticancer activity of Hyal-PEG and a combination of Hyal-PEG with doxorubicin were studied in vitro (morphological analysis of rat glioblastoma 101.8 spheroids) and in vivo (by the survival time of rats after intracerebral transplantation of the tumor and morphological analysis). In the presence of doxorubicin and Hyal-PEG in the culture medium in vitro, spheroids lost their ability to adhere to the substrate and disintegrate into individual cells. Intracerebral transplantation of the tumor tissue with Hyal-PEG did not accelerate glioblastoma growth. The mean survival time for animals receiving transplantation of the tumor alone and in combination with Hyal-PEG was 13 and 20 days, respectively. In one rat with transplanted tumor and Hyal-PEG, this parameter increased by 53%. The survival time of rats receiving systemic therapy with doxorubicin and Hyal-PEG significantly increased (p=0.003). Antitumor effect of therapeutic doses of doxorubicin combined with Hyal-PEG was demonstrated on the model of rat glioblastoma 101.8 in vitro. Hyal-PEG inhibited adhesion of tumor cells, but did not cause their death. Transplantation of Hyal-PEG-treated tumor did not reduce animal survival time. Systemic administration of therapeutic doses of doxorubicin with Hyal-PEG increased survival time of rats with glioblastoma 101.8.


Sujet(s)
Tumeurs du cerveau , Doxorubicine , Glioblastome , Hyaluronoglucosaminidase , Polyéthylène glycols , Animaux , Doxorubicine/pharmacologie , Hyaluronoglucosaminidase/métabolisme , Rats , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Polyéthylène glycols/composition chimique , Polyéthylène glycols/pharmacologie , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Mâle , Lignée cellulaire tumorale , Sphéroïdes de cellules/effets des médicaments et des substances chimiques
18.
Nat Commun ; 15(1): 5871, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38997283

RÉSUMÉ

There are numerous mechanisms by which glioblastoma cells evade immunological detection, underscoring the need for strategic combinatorial treatments to achieve appreciable therapeutic effects. However, developing combination therapies is difficult due to dose-limiting toxicities, blood-brain-barrier, and suppressive tumor microenvironment. Glioblastoma is notoriously devoid of lymphocytes driven in part by a paucity of lymphocyte trafficking factors necessary to prompt their recruitment and activation. Herein, we develop a recombinant adeno-associated virus (AAV) gene therapy that enables focal and stable reconstitution of the tumor microenvironment with C-X-C motif ligand 9 (CXCL9), a powerful call-and-receive chemokine for lymphocytes. By manipulating local chemokine directional guidance, AAV-CXCL9 increases tumor infiltration by cytotoxic lymphocytes, sensitizing glioblastoma to anti-PD-1 immune checkpoint blockade in female preclinical tumor models. These effects are accompanied by immunologic signatures evocative of an inflamed tumor microenvironment. These findings support AAV gene therapy as an adjuvant for reconditioning glioblastoma immunogenicity given its safety profile, tropism, modularity, and off-the-shelf capability.


Sujet(s)
Chimiokine CXCL9 , Dependovirus , Thérapie génétique , Glioblastome , Inhibiteurs de points de contrôle immunitaires , Récepteur-1 de mort cellulaire programmée , Microenvironnement tumoral , Glioblastome/thérapie , Glioblastome/immunologie , Dependovirus/génétique , Microenvironnement tumoral/immunologie , Animaux , Humains , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Chimiokine CXCL9/génétique , Chimiokine CXCL9/immunologie , Souris , Thérapie génétique/méthodes , Femelle , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Lignée cellulaire tumorale , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/thérapie , Vecteurs génétiques/administration et posologie , Vecteurs génétiques/génétique
19.
Sci Rep ; 14(1): 16031, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992201

RÉSUMÉ

O6-methylguanine-DNA methyltransferase (MGMT) has been demonstrated to be an important prognostic and predictive marker in glioblastoma (GBM). To establish a reliable radiomics model based on MRI data to predict the MGMT promoter methylation status of GBM. A total of 183 patients with glioblastoma were included in this retrospective study. The visually accessible Rembrandt images (VASARI) features were extracted for each patient, and a total of 14676 multi-region features were extracted from enhanced, necrotic, "non-enhanced, and edematous" areas on their multiparametric MRI. Twelve individual radiomics models were constructed based on the radiomics features from different subregions and different sequences. Four single-sequence models, three single-region models and the combined radiomics model combining all individual models were constructed. Finally, the predictive performance of adding clinical factors and VASARI characteristics was evaluated. The ComRad model combining all individual radiomics models exhibited the best performance in test set 1 and test set 2, with the area under the receiver operating characteristic curve (AUC) of 0.839 (0.709-0.963) and 0.739 (0.581-0.897), respectively. The results indicated that the radiomics model combining multi-region and multi-parametric MRI features has exhibited promising performance in predicting MGMT methylation status in GBM. The Modeling scheme that combining all individual radiomics models showed best performance among all constructed moels.


Sujet(s)
Tumeurs du cerveau , Méthylation de l'ADN , DNA modification methylases , Enzymes de réparation de l'ADN , Glioblastome , Imagerie par résonance magnétique , Régions promotrices (génétique) , Protéines suppresseurs de tumeurs , Humains , Glioblastome/génétique , Glioblastome/imagerie diagnostique , Glioblastome/anatomopathologie , Enzymes de réparation de l'ADN/génétique , DNA modification methylases/génétique , Protéines suppresseurs de tumeurs/génétique , Imagerie par résonance magnétique/méthodes , Femelle , Mâle , Adulte d'âge moyen , Tumeurs du cerveau/génétique , Tumeurs du cerveau/imagerie diagnostique , Tumeurs du cerveau/anatomopathologie , Études rétrospectives , Adulte , Sujet âgé , Pronostic , Courbe ROC ,
20.
Theranostics ; 14(10): 4107-4126, 2024.
Article de Anglais | MEDLINE | ID: mdl-38994023

RÉSUMÉ

Rationale: The heterogeneity of tumor cells within the glioblastoma (GBM) microenvironment presents a complex challenge in curbing GBM progression. Understanding the specific mechanisms of interaction between different GBM cell subclusters and non-tumor cells is crucial. Methods: In this study, we utilized a comprehensive approach integrating glioma single-cell and spatial transcriptomics. This allowed us to examine the molecular interactions and spatial localization within GBM, focusing on a specific tumor cell subcluster, GBM subcluster 6, and M2-type tumor-associated macrophages (M2 TAMs). Results: Our analysis revealed a significant correlation between a specific tumor cell subcluster, GBM cluster 6, and M2-type TAMs. Further in vitro and in vivo experiments demonstrated the specific regulatory role of the CEBPB transcriptional network in GBM subcluster 6, which governs its tumorigenicity, recruitment of M2 TAMs, and polarization. This regulation involves molecules such as MCP1 for macrophage recruitment and the SPP1-Integrin αvß1-Akt signaling pathway for M2 polarization. Conclusion: Our findings not only deepen our understanding of the formation of M2 TAMs, particularly highlighting the differential roles played by heterogeneous cells within GBM in this process, but also provided new insights for effectively controlling the malignant progression of GBM.


Sujet(s)
Protéine bêta de liaison aux séquences stimulatrices de type CCAAT , Glioblastome , Microenvironnement tumoral , Macrophages associés aux tumeurs , Glioblastome/anatomopathologie , Glioblastome/métabolisme , Glioblastome/génétique , Humains , Protéine bêta de liaison aux séquences stimulatrices de type CCAAT/métabolisme , Protéine bêta de liaison aux séquences stimulatrices de type CCAAT/génétique , Animaux , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/immunologie , Souris , Lignée cellulaire tumorale , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/génétique , Régulation de l'expression des gènes tumoraux , Transduction du signal , Macrophages/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...