Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 6.329
Filtrer
1.
Nagoya J Med Sci ; 86(2): 304-313, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38962412

RÉSUMÉ

Radiotherapy combined with temozolomide (TMZ+RT) is the primary treatment for high-grade glioma. TMZ is classified as a moderate emetic risk agent and, thus, supportive care for nausea and vomiting is important. In Nagoya University Hospital, all patients are treated with a 5-hydroxy-tryptamine 3 receptor antagonist (5-HT3RA) for the first 3 days. The daily administration of 5-HT3RA is resumed after the 4th day based on the condition of patients during TMZ+RT. Therefore, the present study investigated risk factors for nausea and vomiting in patients requiring the daily administration of 5-HT3RA. Patients with high-grade glioma who received TMZ+RT between January 2014 and December 2019 at our hospital were included. Patients were divided into two groups: a control group (patients who did not resume 5-HT3RA) and resuming 5-HT3RA group (patients who resumed 5-HT3RA after the 4th day), and both groups were compared to identify risk factors for nausea and vomiting during TMZ+RT. There were 78 patients in the control group (68%) and 36 in the resuming 5-HT3RA group (32%). A multivariate analysis of patient backgrounds in the two groups identified age <18 years, PS 2 or more, and occipital lobe tumors as risk factors for nausea and vomiting. Nausea and vomiting were attenuated in 30 patients (83%) in the resuming 5-HT3RA group following the resumption of 5-HT3RA. The results obtained highlight the importance of extracting patients with these risk factors before the initiation of therapy and the early resumption or daily administration of 5-HT3RA according to the condition of each patient.


Sujet(s)
Gliome , Nausée , Antagonistes des récepteurs 5-HT3 de la sérotonine , Témozolomide , Vomissement , Humains , Témozolomide/usage thérapeutique , Témozolomide/administration et posologie , Témozolomide/effets indésirables , Mâle , Antagonistes des récepteurs 5-HT3 de la sérotonine/usage thérapeutique , Antagonistes des récepteurs 5-HT3 de la sérotonine/administration et posologie , Femelle , Vomissement/induit chimiquement , Vomissement/traitement médicamenteux , Adulte d'âge moyen , Gliome/traitement médicamenteux , Gliome/radiothérapie , Facteurs de risque , Sujet âgé , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/radiothérapie , Adulte , Antinéoplasiques alcoylants/usage thérapeutique , Antinéoplasiques alcoylants/effets indésirables , Antinéoplasiques alcoylants/administration et posologie , Chimioradiothérapie/effets indésirables , Chimioradiothérapie/méthodes
2.
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
3.
Cancer Med ; 13(13): e7385, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38970256

RÉSUMÉ

BACKGROUND: Pamiparib is a potent, selective, poly (ADP-ribose) polymerase 1/2 inhibitor that demonstrates synthetic lethality in cells with breast cancer susceptibility gene mutations or other homologous recombination deficiency. This two-stage phase 1b study (NCT03150810) assessed pamiparib in combination with temozolomide (TMZ) in adult patients with histologically confirmed locally advanced and metastatic solid tumors. METHODS: Oral pamiparib 60 mg was administered twice daily. During the dose-escalation stage, increasing doses of TMZ (40-120 mg once daily pulsed or 20-40 mg once daily continuous) were administered to determine the recommended dose to be administered in the dose-expansion stage. The primary objectives were to determine safety and tolerability, maximum tolerated/administered dose, recommended phase 2 dose and schedule, and antitumor activity of pamiparib in combination with TMZ. Pharmacokinetics of pamiparib and TMZ and biomarkers were also assessed. RESULTS: Across stages, 139 patients were treated (dose escalation, n = 66; dose expansion, n = 73). The maximum tolerated dose of TMZ, which was administered during dose expansion, was 7-day pulsed 60 mg once daily. The most common treatment-emergent adverse events (TEAEs) were anemia (dose escalation, 56.1%; dose expansion, 63.0%), nausea (dose escalation, 54.5%; dose expansion, 49.3%), and fatigue (dose escalation, 48.5%; dose expansion, 47.9%). In the dose-escalation stage, four patients experienced dose-limiting toxicities (three neutropenia and one neutrophil count decreased). No TEAEs considered to be related to study drug treatment resulted in death. Antitumor activity was modest, indicated by confirmed overall response rate (dose escalation, 13.8%; dose expansion, 11.6%), median progression-free survival (3.7 and 2.8 months), and median overall survival (10.5 and 9.2 months). Administration of combination therapy did not notably impact pamiparib or TMZ pharmacokinetics. CONCLUSIONS: Pamiparib in combination with TMZ had a manageable safety profile. Further investigation of the efficacy of this combination in tumor types with specific DNA damage repair deficiencies is warranted.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Benzimidazoles , Dose maximale tolérée , Tumeurs , Témozolomide , Humains , Témozolomide/administration et posologie , Témozolomide/pharmacocinétique , Témozolomide/effets indésirables , Témozolomide/usage thérapeutique , Femelle , Adulte d'âge moyen , Mâle , Sujet âgé , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Adulte , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Benzimidazoles/administration et posologie , Benzimidazoles/pharmacocinétique , Benzimidazoles/effets indésirables , Benzimidazoles/usage thérapeutique , Sujet âgé de 80 ans ou plus , Inhibiteurs de poly(ADP-ribose) polymérases/administration et posologie , Inhibiteurs de poly(ADP-ribose) polymérases/effets indésirables , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacocinétique , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Calendrier d'administration des médicaments , Fluorènes
4.
Cancer Res Commun ; 4(6): 1566-1580, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38837899

RÉSUMÉ

Glioblastoma (GBM) is the deadliest adult brain cancer. Under the current standard of care, almost all patients succumb to the disease and novel treatments are urgently needed. Recognizing that GBMs are addicted to cholesterol, past clinical trials have repurposed statins against GBM but failed. The purpose of this study was to test whether treatments that upregulate the cholesterol biosynthesis pathway in GBM would generate a metabolic vulnerability that can be exploited using statins and to determine the underlying mechanisms.Effects of radiotherapy and temozolomide or dopamine receptor antagonists on the mevalonate pathway in GBM were assessed in vitro and in vivo. The impact of statins on self-renewal of glioma stem cells and median survival was studied. Branches of the mevalonate pathway were probed to identify relevant effector proteins.Cells surviving combination treatments that converge in activating the immediate early response, universally upregulated the mevalonate pathway and increased stemness of GBM cells through activation of the Rho-GTPase Rac-1. Activation of the mevalonate pathway and Rac-1 was inhibited by statins, which led to improved survival in mouse models of glioblastoma when combined with radiation and drugs that target the glioma stem cell pool and plasticity of glioma cells.We conclude that a combination of dopamine receptor antagonists and statins could potentially improve radiotherapy outcome and warrants further investigation. SIGNIFICANCE: Combination therapies that activate the mevalonate pathway in GBM cells after sublethal treatment enhance self-renewal and migratory capacity through Rac-1 activation, which creates a metabolic vulnerability that can be further potentially exploited using statins.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Acide mévalonique , Témozolomide , Protéine G rac1 , Glioblastome/traitement médicamenteux , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Acide mévalonique/métabolisme , Humains , Animaux , Protéine G rac1/métabolisme , Protéine G rac1/antagonistes et inhibiteurs , Souris , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Lignée cellulaire tumorale , Témozolomide/pharmacologie , Témozolomide/usage thérapeutique , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/pharmacologie , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/usage thérapeutique , Récidive tumorale locale/traitement médicamenteux , Récidive tumorale locale/anatomopathologie , Tests d'activité antitumorale sur modèle de xénogreffe , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Antagonistes de la dopamine/pharmacologie
5.
Genes (Basel) ; 15(6)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38927654

RÉSUMÉ

Glioblastoma multiforme (GBM)is the most common and aggressive primary brain tumor. Although temozolomide (TMZ)-based radiochemotherapy improves overall GBM patients' survival, it also increases the frequency of false positive post-treatment magnetic resonance imaging (MRI) assessments for tumor progression. Pseudo-progression (PsP) is a treatment-related reaction with an increased contrast-enhancing lesion size at the tumor site or resection margins miming tumor recurrence on MRI. The accurate and reliable prognostication of GBM progression is urgently needed in the clinical management of GBM patients. Clinical data analysis indicates that the patients with PsP had superior overall and progression-free survival rates. In this study, we aimed to develop a prognostic model to evaluate the tumor progression potential of GBM patients following standard therapies. We applied a dictionary learning scheme to obtain imaging features of GBM patients with PsP or true tumor progression (TTP) from the Wake dataset. Based on these radiographic features, we conducted a radiogenomics analysis to identify the significantly associated genes. These significantly associated genes were used as features to construct a 2YS (2-year survival rate) logistic regression model. GBM patients were classified into low- and high-survival risk groups based on the individual 2YS scores derived from this model. We tested our model using an independent The Cancer Genome Atlas Program (TCGA) dataset and found that 2YS scores were significantly associated with the patient's overall survival. We used two cohorts of the TCGA data to train and test our model. Our results show that the 2YS scores-based classification results from the training and testing TCGA datasets were significantly associated with the overall survival of patients. We also analyzed the survival prediction ability of other clinical factors (gender, age, KPS (Karnofsky performance status), normal cell ratio) and found that these factors were unrelated or weakly correlated with patients' survival. Overall, our studies have demonstrated the effectiveness and robustness of the 2YS model in predicting the clinical outcomes of GBM patients after standard therapies.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Imagerie par résonance magnétique , Humains , Glioblastome/génétique , Glioblastome/imagerie diagnostique , Glioblastome/anatomopathologie , Glioblastome/mortalité , Tumeurs du cerveau/génétique , Tumeurs du cerveau/imagerie diagnostique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/mortalité , Mâle , Femelle , Imagerie par résonance magnétique/méthodes , Adulte d'âge moyen , Pronostic , Adulte , Sujet âgé , Évolution de la maladie , Témozolomide/usage thérapeutique , Génomique/méthodes , Taux de survie , Pertinence clinique
6.
Cell Death Dis ; 15(6): 454, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38937431

RÉSUMÉ

Despite being mutated in 92% of TP53 mutant cancers, how mutations on p53 isoforms affect their activities remain largely unknown. Therefore, exploring the effect of mutations on p53 isoforms activities is a critical, albeit unexplored area in the p53 field. In this article, we report for the first time a mutant Δ133p53α-specific pathway which increases IL4I1 and IDO1 expression and activates AHR, a tumor-promoting mechanism. Accordingly, while WT Δ133p53α reduces apoptosis to promote DNA repair, mutant R273H also reduces apoptosis but fails to maintain genomic stability, increasing the risks of accumulation of mutations and tumor's deriving towards a more aggressive phenotype. Furthermore, using 2D and 3D spheroids culture, we show that WT Δ133p53α reduces cell proliferation, EMT, and invasion, while the mutant Δ133p53α R273H enhances all three processes, confirming its oncogenic potential and strongly suggesting a similar in vivo activity. Importantly, the effects on cell growth and invasion are independent of mutant full-length p53α, indicating that these activities are actively carried by mutant Δ133p53α R273H. Furthermore, both WT and mutant Δ133p53α reduce cellular senescence in a senescence inducer-dependent manner (temozolomide or radiation) because they regulate different senescence-associated target genes. Hence, WT Δ133p53α rescues temozolomide-induced but not radiation-induced senescence, while mutant Δ133p53α R273H rescues radiation-induced but not temozolomide-induced senescence. Lastly, we determined that IL4I1, IDO1, and AHR are significantly higher in GBMs compared to low-grade gliomas. Importantly, high expression of all three genes in LGG and IL4I1 in GBM is significantly associated with poorer patients' survival, confirming the clinical relevance of this pathway in glioblastomas. These data show that, compared to WT Δ133p53α, R273H mutation reorientates its activities toward carcinogenesis and activates the oncogenic IL4I1/IDO1/AHR pathway, a potential prognostic marker and therapeutic target in GBM by combining drugs specifically modulating Δ133p53α expression and IDO1/Il4I1/AHR inhibitors.


Sujet(s)
Prolifération cellulaire , Vieillissement de la cellule , Glioblastome , Mutation , Protéine p53 suppresseur de tumeur , Glioblastome/génétique , Glioblastome/anatomopathologie , Glioblastome/métabolisme , Humains , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Vieillissement de la cellule/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mutation/génétique , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Témozolomide/pharmacologie , Témozolomide/usage thérapeutique
7.
Sci Adv ; 10(23): eadj7706, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38848360

RÉSUMÉ

Poor prognosis and drug resistance in glioblastoma (GBM) can result from cellular heterogeneity and treatment-induced shifts in phenotypic states of tumor cells, including dedifferentiation into glioma stem-like cells (GSCs). This rare tumorigenic cell subpopulation resists temozolomide, undergoes proneural-to-mesenchymal transition (PMT) to evade therapy, and drives recurrence. Through inference of transcriptional regulatory networks (TRNs) of patient-derived GSCs (PD-GSCs) at single-cell resolution, we demonstrate how the topology of transcription factor interaction networks drives distinct trajectories of cell-state transitions in PD-GSCs resistant or susceptible to cytotoxic drug treatment. By experimentally testing predictions based on TRN simulations, we show that drug treatment drives surviving PD-GSCs along a trajectory of intermediate states, exposing vulnerability to potentiated killing by siRNA or a second drug targeting treatment-induced transcriptional programs governing nongenetic cell plasticity. Our findings demonstrate an approach to uncover TRN topology and use it to rationally predict combinatorial treatments that disrupt acquired resistance in GBM.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Régulation de l'expression des gènes tumoraux , Réseaux de régulation génique , Gliome , Cellules souches tumorales , Humains , Cellules souches tumorales/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/anatomopathologie , Résistance aux médicaments antinéoplasiques/génétique , Gliome/génétique , Gliome/anatomopathologie , Gliome/métabolisme , Gliome/traitement médicamenteux , Témozolomide/pharmacologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/traitement médicamenteux , Lignée cellulaire tumorale , Glioblastome/génétique , Glioblastome/anatomopathologie , Glioblastome/métabolisme , Glioblastome/traitement médicamenteux
8.
Int J Mol Sci ; 25(11)2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38892466

RÉSUMÉ

Glioblastoma (GBM) is the most common primary malignant brain tumor in adults, with few effective treatments. EGFR alterations, including expression of the truncated variant EGFRvIII, are among the most frequent genomic changes in these tumors. EGFRvIII is known to preferentially signal through STAT5 for oncogenic activation in GBM, yet targeting EGFRvIII has yielded limited clinical success to date. In this study, we employed patient-derived xenograft (PDX) models expressing EGFRvIII to determine the key points of therapeutic vulnerability within the EGFRvIII-STAT5 signaling axis in GBM. Our findings reveal that exogenous expression of paralogs STAT5A and STAT5B augments cell proliferation and that inhibition of STAT5 phosphorylation in vivo improves overall survival in combination with temozolomide (TMZ). STAT5 phosphorylation is independent of JAK1 and JAK2 signaling, instead requiring Src family kinase (SFK) activity. Saracatinib, an SFK inhibitor, attenuates phosphorylation of STAT5 and preferentially sensitizes EGFRvIII+ GBM cells to undergo apoptotic cell death relative to wild-type EGFR. Constitutively active STAT5A or STAT5B mitigates saracatinib sensitivity in EGFRvIII+ cells. In vivo, saracatinib treatment decreased survival in mice bearing EGFR WT tumors compared to the control, yet in EGFRvIII+ tumors, treatment with saracatinib in combination with TMZ preferentially improves survival.


Sujet(s)
Benzodioxoles , Prolifération cellulaire , Récepteurs ErbB , Glioblastome , Quinazolines , Facteur de transcription STAT-5 , Témozolomide , Facteur de transcription STAT-5/métabolisme , Glioblastome/traitement médicamenteux , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Glioblastome/génétique , Humains , Animaux , Quinazolines/pharmacologie , Quinazolines/usage thérapeutique , Benzodioxoles/pharmacologie , Benzodioxoles/usage thérapeutique , Souris , Récepteurs ErbB/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Témozolomide/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Transduction du signal/effets des médicaments et des substances chimiques , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/génétique , Apoptose/effets des médicaments et des substances chimiques , src-Family kinases/métabolisme , Protéines suppresseurs de tumeurs
9.
Cells ; 13(11)2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38891029

RÉSUMÉ

Glioblastoma Multiforme (GBM) is an aggressive brain tumor with a high mortality rate. Direct reprogramming of glial cells to different cell lineages, such as induced neural stem cells (iNSCs) and induced neurons (iNeurons), provides genetic tools to manipulate a cell's fate as a potential therapy for neurological diseases. NeuroD1 (ND1) is a master transcriptional factor for neurogenesis and it promotes neuronal differentiation. In the present study, we tested the hypothesis that the expression of ND1 in GBM cells can force them to differentiate toward post-mitotic neurons and halt GBM tumor progression. In cultured human GBM cell lines, including LN229, U87, and U373 as temozolomide (TMZ)-sensitive and T98G as TMZ-resistant cells, the neuronal lineage conversion was induced by an adeno-associated virus (AAV) package carrying ND1. Twenty-one days after AAV-ND1 transduction, ND1-expressing cells displayed neuronal markers MAP2, TUJ1, and NeuN. The ND1-induced transdifferentiation was regulated by Wnt signaling and markedly enhanced under a hypoxic condition (2% O2 vs. 21% O2). ND1-expressing GBM cultures had fewer BrdU-positive proliferating cells compared to vector control cultures. Increased cell death was visualized by TUNEL staining, and reduced migrative activity was demonstrated in the wound-healing test after ND1 reprogramming in both TMZ-sensitive and -resistant GBM cells. In a striking contrast to cancer cells, converted cells expressed the anti-tumor gene p53. In an orthotopical GBM mouse model, AAV-ND1-reprogrammed U373 cells were transplanted into the fornix of the cyclosporine-immunocompromised C57BL/6 mouse brain. Compared to control GBM cell-formed tumors, cells from ND1-reprogrammed cultures formed smaller tumors and expressed neuronal markers such as TUJ1 in the brain. Thus, reprogramming using a single-factor ND1 overcame drug resistance, converting malignant cells of heterogeneous GBM cells to normal neuron-like cells in vitro and in vivo. These novel observations warrant further research using patient-derived GBM cells and patient-derived xenograft (PDX) models as a potentially effective treatment for a deadly brain cancer and likely other astrocytoma tumors.


Sujet(s)
Reprogrammation cellulaire , Glioblastome , Neurones , Glioblastome/anatomopathologie , Glioblastome/génétique , Glioblastome/métabolisme , Humains , Animaux , Lignée cellulaire tumorale , Neurones/métabolisme , Neurones/effets des médicaments et des substances chimiques , Souris , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/métabolisme , Témozolomide/pharmacologie , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique
10.
Oncol Res ; 32(6): 1037-1045, 2024.
Article de Anglais | MEDLINE | ID: mdl-38827324

RÉSUMÉ

Background: The dysregulation of Isocitrate dehydrogenase (IDH) and the subsequent production of 2-Hydroxyglutrate (2HG) may alter the expression of epigenetic proteins in Grade 4 astrocytoma. The interplay mechanism between IDH, O-6-methylguanine-DNA methyltransferase (MGMT)-promoter methylation, and protein methyltransferase proteins-5 (PRMT5) activity, with tumor progression has never been described. Methods: A retrospective cohort of 34 patients with G4 astrocytoma is classified into IDH-mutant and IDH-wildtype tumors. Both groups were tested for MGMT-promoter methylation and PRMT5 through methylation-specific and gene expression PCR analysis. Inter-cohort statistical significance was evaluated. Results: Both IDH-mutant WHO grade 4 astrocytomas (n = 22, 64.7%) and IDH-wildtype glioblastomas (n = 12, 35.3%) had upregulated PRMT5 gene expression except in one case. Out of the 22 IDH-mutant tumors, 10 (45.5%) tumors showed MGMT-promoter methylation and 12 (54.5%) tumors had unmethylated MGMT. All IDH-wildtype tumors had unmethylated MGMT. There was a statistically significant relationship between MGMT-promoter methylation and IDH in G4 astrocytoma (p-value = 0.006). Statistically significant differences in progression-free survival (PFS) were also observed among all G4 astrocytomas that expressed PRMT5 and received either temozolomide (TMZ) or TMZ plus other chemotherapies, regardless of their IDH or MGMT-methylation status (p-value=0.0014). Specifically, IDH-mutant tumors that had upregulated PRMT5 activity and MGMT-promoter methylation, who received only TMZ, have exhibited longer PFS. Conclusions: The relationship between PRMT5, MGMT-promoter, and IDH is not tri-directional. However, accumulation of D2-hydroxyglutarate (2-HG), which partially activates 2-OG-dependent deoxygenase, may not affect their activities. In IDH-wildtype glioblastomas, the 2HG-2OG pathway is typically inactive, leading to PRMT5 upregulation. TMZ alone, compared to TMZ-plus, can increase PFS in upregulated PRMT5 tumors. Thus, using a PRMT5 inhibitor in G4 astrocytomas may help in tumor regression.


Sujet(s)
Astrocytome , Méthylation de l'ADN , DNA modification methylases , Enzymes de réparation de l'ADN , Évolution de la maladie , Isocitrate dehydrogenases , Mutation , Régions promotrices (génétique) , Protein-arginine N-methyltransferases , Protéines suppresseurs de tumeurs , Humains , Protein-arginine N-methyltransferases/génétique , Protein-arginine N-methyltransferases/métabolisme , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme , Enzymes de réparation de l'ADN/génétique , Enzymes de réparation de l'ADN/métabolisme , DNA modification methylases/génétique , DNA modification methylases/métabolisme , Isocitrate dehydrogenases/génétique , Mâle , Femelle , Astrocytome/génétique , Astrocytome/anatomopathologie , Adulte d'âge moyen , Adulte , Études rétrospectives , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Grading des tumeurs , Sujet âgé , Témozolomide/usage thérapeutique , Témozolomide/pharmacologie , Régulation de l'expression des gènes tumoraux
11.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 178-186, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38836664

RÉSUMÉ

This study aimed to explore the regulatory effects and associated mechanisms of adiponectin on apoptosis and proliferation in the LN18 glioma cell line through the AMPK and Akt signaling pathways. Additionally, we sought to elucidate the impact of adiponectin on the chemosensitivity of the LN18 glioma cell line to temozolomide (TMZ). The proliferation rate of glioma cells treated with adiponectin was assessed using the cholecystokinin (CCK8) assay. The Western blot analysis was employed to assess the expression of p-Akt, p-AMPK, p-mTOR, cleaved caspase3, Bax, Cyclin D1, and Cyclin B1 following adiponectin treatment. Cell apoptosis was quantified using AnnexinV/PI flow cytometry, while changes in the cell cycle were detected using PI staining flow cytometry. The findings revealed that adiponectin upregulates p-AMPK expression and downregulates p-mTOR expression in the PTEN wild-type glioma cell line LN18, with no discernible effect on p-Akt expression. Moreover, adiponectin inhibits the proliferation rate of the PTEN wild-type glioma cell line LN18, enhances the expression of cleaved caspase3 and Bax, and significantly elevates the apoptosis rate, as evidenced by AnnexinV/PI flow cytometry. Adiponectin was observed to suppress the expression of Cyclin D1 and Cyclin B1, increase the number of cells in the G1 phase, and promote autophagy. Additionally, adiponectin augments the expression of Beclin1 and the ratio of LC3II/I in the PTEN wild-type glioma cell line LN18, while decreasing p62 expression. In conclusion, this study posits that adiponectin holds therapeutic promise for glioma treatment. Furthermore, adiponectin enhances the inhibitory effect of TMZ on the proliferation rate of LN18 cells when treated with 0.1 mM and 1 mM TMZ. These results collectively suggest that adiponectin impedes proliferation, encourages apoptosis and autophagy in the LN18 glioma cell line, and heightens its sensitivity to the chemotherapeutic drug TMZ.


Sujet(s)
Adiponectine , Apoptose , Autophagie , Prolifération cellulaire , Gliome , Témozolomide , Adiponectine/métabolisme , Adiponectine/pharmacologie , Adiponectine/génétique , Apoptose/effets des médicaments et des substances chimiques , Humains , Gliome/anatomopathologie , Gliome/métabolisme , Gliome/traitement médicamenteux , Gliome/génétique , Autophagie/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Témozolomide/pharmacologie , Protéines proto-oncogènes c-akt/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , AMP-Activated Protein Kinases/métabolisme
12.
Nat Commun ; 15(1): 5294, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38906885

RÉSUMÉ

Determining the balance between DNA double strand break repair (DSBR) pathways is essential for understanding treatment response in cancer. We report a method for simultaneously measuring non-homologous end joining (NHEJ), homologous recombination (HR), and microhomology-mediated end joining (MMEJ). Using this method, we show that patient-derived glioblastoma (GBM) samples with acquired temozolomide (TMZ) resistance display elevated HR and MMEJ activity, suggesting that these pathways contribute to treatment resistance. We screen clinically relevant small molecules for DSBR inhibition with the aim of identifying improved GBM combination therapy regimens. We identify the ATM kinase inhibitor, AZD1390, as a potent dual HR/MMEJ inhibitor that suppresses radiation-induced phosphorylation of DSBR proteins, blocks DSB end resection, and enhances the cytotoxic effects of TMZ in treatment-naïve and treatment-resistant GBMs with TP53 mutation. We further show that a combination of G2/M checkpoint deficiency and reliance upon ATM-dependent DSBR renders TP53 mutant GBMs hypersensitive to TMZ/AZD1390 and radiation/AZD1390 combinations. This report identifies ATM-dependent HR and MMEJ as targetable resistance mechanisms in TP53-mutant GBM and establishes an approach for simultaneously measuring multiple DSBR pathways in treatment selection and oncology research.


Sujet(s)
Protéines mutées dans l'ataxie-télangiectasie , Cassures double-brin de l'ADN , Glioblastome , Témozolomide , Protéine p53 suppresseur de tumeur , Humains , Protéines mutées dans l'ataxie-télangiectasie/métabolisme , Protéines mutées dans l'ataxie-télangiectasie/antagonistes et inhibiteurs , Protéines mutées dans l'ataxie-télangiectasie/génétique , Glioblastome/génétique , Glioblastome/traitement médicamenteux , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Cassures double-brin de l'ADN/effets des médicaments et des substances chimiques , Témozolomide/pharmacologie , Lignée cellulaire tumorale , Mutation , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Réparation de l'ADN/effets des médicaments et des substances chimiques , Tumeurs du cerveau/génétique , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Animaux , Réparation de l'ADN par jonction d'extrémités/effets des médicaments et des substances chimiques , Souris , Phosphorylation/effets des médicaments et des substances chimiques
13.
Sci Rep ; 14(1): 14341, 2024 06 21.
Article de Anglais | MEDLINE | ID: mdl-38906916

RÉSUMÉ

Glioblastoma (GBM) is a highly aggressive and deadly brain cancer. Temozolomide (TMZ) is the standard chemotherapeutic agent for GBM, but the majority of patients experience recurrence and invasion of tumor cells. We investigated whether TMZ treatment of GBM cells regulates matrix metalloproteinases (MMPs), which have the main function to promote tumor cell invasion. TMZ effectively killed GL261, U343, and U87MG cells at a concentration of 500 µM, and surviving cells upregulated MMP9 expression and its activity but not those of MMP2. TMZ also elevated levels of MMP9 mRNA and MMP9 promoter activity. Subcutaneous graft tumors survived from TMZ treatment also exhibited increased expression of MMP9 and enhanced gelatinolytic activity. TMZ-mediated MMP9 upregulation was specifically mediated through the phosphorylation of p38 and JNK. This then stimulates AP-1 activity through the upregulation of c-Fos and c-Jun. Inhibition of the p38, JNK, or both pathways counteracted the TMZ-induced upregulation of MMP9 and AP-1. This study proposes a potential adverse effect of TMZ treatment for GBM: upregulation of MMP9 expression potentially associated with increased invasion and poor prognosis. This study also provides valuable insights into the molecular mechanisms by which TMZ treatment leads to increased MMP9 expression in GBM cells.


Sujet(s)
Régulation de l'expression des gènes tumoraux , Glioblastome , Matrix metalloproteinase 9 , Témozolomide , p38 Mitogen-Activated Protein Kinases , Témozolomide/pharmacologie , Glioblastome/traitement médicamenteux , Glioblastome/métabolisme , Glioblastome/génétique , Glioblastome/anatomopathologie , Matrix metalloproteinase 9/métabolisme , Matrix metalloproteinase 9/génétique , Humains , p38 Mitogen-Activated Protein Kinases/métabolisme , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Antinéoplasiques alcoylants/pharmacologie , Animaux , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Facteur de transcription AP-1/métabolisme , Régulation positive/effets des médicaments et des substances chimiques , Souris
14.
BMC Cancer ; 24(1): 754, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38907215

RÉSUMÉ

PURPOSE: Diffuse midline glioma (DMG), H3 K27M-mutant is a type of diffuse high-grade glioma that occurs in the brain midline carrying an extremely poor prognosis under the best efforts of surgery, radiation, and other therapies. For better therapy, we explored the efficacy and toxicity of a novel therapy that combines apatinib and temozolomide in DMG. METHODS: A retrospective analysis of 32 patients with DMG who underwent apatinib plus temozolomide treatment was performed. Apatinib was given 500 mg in adults, 250 mg in pediatric patients once daily. Temozolomide was administered at 200 mg/m2/d according to the standard 5/28 days regimen. The main clinical data included basic information of patients, radiological and pathological characteristics of tumors, treatment, adverse reactions, prognosis. RESULTS: The objective response rate was 24.1%, and the disease control rate was 79.3%. The median PFS of all patients was 5.8 months, and median OS was 10.3 months. A total of 236 cycles of treatment were available for safety assessment and the toxicity of the combination therapy was relatively well tolerated. The most common grade 3 toxicities were myelosuppression including leukopenia (5.08%), neutropenia (4.24%), lymphopenia (2.12%), thrombocytopenia (1.69%) and anemia (1.27%). Grade 4 toxicities included neutropenia (2.12%), thrombocytopenia (2.12%) and proteinuria (1.69%). All the adverse events were relieved after symptomatic treatment or dose reduction. CONCLUSIONS: Apatinib plus temozolomide could be an effective regimen with manageable toxicities and favorable efficacy and may outperform temozolomide monotherapy, particularly in newly diagnosed adults with tumors located outside the pons. The novel therapy deserves further investigation in adult DMG patients.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Tumeurs du cerveau , Gliome , Pyridines , Témozolomide , Humains , Témozolomide/administration et posologie , Témozolomide/usage thérapeutique , Témozolomide/effets indésirables , Femelle , Mâle , Adulte , Pyridines/administration et posologie , Pyridines/effets indésirables , Pyridines/usage thérapeutique , Gliome/traitement médicamenteux , Gliome/anatomopathologie , Adolescent , Études rétrospectives , Enfant , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Jeune adulte , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Enfant d'âge préscolaire , Adulte d'âge moyen , Résultat thérapeutique
15.
Redox Biol ; 73: 103220, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38838551

RÉSUMÉ

Temozolomide (TMZ) is a widely utilized chemotherapy treatment for patients with glioblastoma (GBM), although drug resistance constitutes a major therapeutic hurdle. Emerging evidence suggests that ferroptosis-mediated therapy could offer an appropriate alternative treatment option against cancer cells that are resistant to certain drugs. However, recurrent gliomas display robust ferroptosis resistance, although the precise mechanism of resistance remains elusive. In the present work, we report that proline rich protein 11 (PRR11) depletion significantly sensitizes GBM cells to TMZ by inducing ferroptosis. Mechanistically, PRR11 directly binds to and stabilizes dihydroorotate dehydrogenase (DHODH), which leads to glioma ferroptosis-resistant in a DHODH-dependent manner in vivo and in vitro. Furthermore, PRR11 inhibits HERC4 and DHODH binding, by suppressing the recruitment of E3 ubiquitin ligase HERC4 and polyubiquitination degradation of DHODH at the K306 site, which maintains DHODH protein stability. Importantly, downregulated PRR11 increases lipid peroxidation and alters DHODH-mediated mitochondrial morphology, thereby promoting ferroptosis and increasing TMZ chemotherapy sensitivity. In conclusion, our results reveal a mechanism via which PRR11 drives ferroptosis resistance and identifies ferroptosis induction and TMZ as an attractive combined therapeutic strategy for GBM.


Sujet(s)
Dihydroorotate dehydrogenase , Résistance aux médicaments antinéoplasiques , Ferroptose , Glioblastome , Témozolomide , Humains , Ferroptose/effets des médicaments et des substances chimiques , Ferroptose/génétique , Glioblastome/métabolisme , Glioblastome/traitement médicamenteux , Glioblastome/génétique , Glioblastome/anatomopathologie , Témozolomide/pharmacologie , Résistance aux médicaments antinéoplasiques/génétique , Lignée cellulaire tumorale , Souris , Dihydroorotate dehydrogenase/métabolisme , Animaux , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/génétique
16.
Anticancer Res ; 44(7): 2943-2952, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38925834

RÉSUMÉ

BACKGROUND/AIM: Glioblastoma is an incurable cancer with limited treatment options and a low survival rate. Temozolomide is the standard marketed small-molecule agent for glioblastoma therapy; therefore, we aimed to find new drugs among the marketed medicines for brain diseases because of their cerebral migratory property and found lomerizine, used for the treatment of migraine. MATERIALS AND METHODS: We evaluated the effect of lomerizine and its metabolites against U251 glioblastoma cells and temozolomide-resistant cells, T98G and GB-1, caused by the expression of O(6)-methylguanine-DNA methyltransferase or P-glycoprotein, compared with temozolomide, and combined with it. The mechanism of action was investigated using inhibitors of necrosis or apoptosis. RESULTS: Lomerizine and its metabolite (M6) inhibited the proliferation of glioblastoma cells with greater potency and efficacy than temozolomide, including against temozolomide-resistant cells. The effects of lomerizine and M6 on glioblastoma were mainly attributed to the inhibition of proliferation because cells were not rescued by cell death inhibitors, such as necrosis or apoptosis inhibitors, although they were slightly rescued by necrostatin-1. Additionally, lomerizine and M6 combined with temozolomide were more effective at inhibiting the proliferation of U251 and GB-1 cells at some doses than single treatments. CONCLUSION: Lomerizine has been used for migraine treatment because of its brain-penetrating properties without serious side-effects; thus, it might potentially be expected to be used alone for glioblastoma, including temozolomide-resistant glioblastoma, or in combination with temozolomide.


Sujet(s)
Apoptose , Prolifération cellulaire , Glioblastome , Témozolomide , Humains , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Glioblastome/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Témozolomide/pharmacologie , Pipérazines/pharmacologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Dacarbazine/pharmacologie , Dacarbazine/analogues et dérivés
17.
Lancet Oncol ; 25(7): 922-932, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38936379

RÉSUMÉ

BACKGROUND: Neuroblastoma is the most common extracranial solid tumour in children. Relapsed or refractory neuroblastoma is associated with a poor outcome. We assessed the combination of irinotecan-temozolomide and dasatinib-rapamycin (RIST) in patients with relapsed or refractory neuroblastoma. METHODS: The multicentre, open-label, randomised, controlled, phase 2, RIST-rNB-2011 trial recruited from 40 paediatric oncology centres in Germany and Austria. Patients aged 1-25 years with high-risk relapsed (defined as recurrence of all stage IV and MYCN amplification stages, after response to treatment) or refractory (progressive disease during primary treatment) neuroblastoma, with Lansky and Karnofsky performance status at least 50%, were assigned (1:1) to RIST (RIST group) or irinotecan-temozolomide (control group) by block randomisation, stratified by MYCN status. We compared RIST (oral rapamycin [loading 3 mg/m2 on day 1, maintenance 1 mg/m2 on days 2-4] and oral dasatinib [2 mg/kg per day] for 4 days with 3 days off, followed by intravenous irinotecan [50 mg/m2 per day] and oral temozolomide [150 mg/m2 per day] for 5 days with 2 days off; one course each of rapamycin-dasatinib and irinotecan-temozolomide for four cycles over 8 weeks, then two courses of rapamycin-dasatinib followed by one course of irinotecan-temozolomide for 12 weeks) with irinotecan-temozolomide alone (with identical dosing as experimental group). The primary endpoint of progression-free survival was analysed in all eligible patients who received at least one course of therapy. The safety population consisted of all patients who received at least one course of therapy and had at least one post-baseline safety assessment. This trial is registered at ClinicalTrials.gov, NCT01467986, and is closed to accrual. FINDINGS: Between Aug 26, 2013, and Sept 21, 2020, 129 patients were randomly assigned to the RIST group (n=63) or control group (n=66). Median age was 5·4 years (IQR 3·7-8·1). 124 patients (78 [63%] male and 46 [37%] female) were included in the efficacy analysis. At a median follow-up of 72 months (IQR 31-88), the median progression-free survival was 11 months (95% CI 7-17) in the RIST group and 5 months (2-8) in the control group (hazard ratio 0·62, one-sided 90% CI 0·81; p=0·019). Median progression-free survival in patients with amplified MYCN (n=48) was 6 months (95% CI 4-24) in the RIST group versus 2 months (2-5) in the control group (HR 0·45 [95% CI 0·24-0·84], p=0·012); median progression-free survival in patients without amplified MYCN (n=76) was 14 months (95% CI 9-7) in the RIST group versus 8 months (4-15) in the control group (HR 0·84 [95% CI 0·51-1·38], p=0·49). The most common grade 3 or worse adverse events were neutropenia (54 [81%] of 67 patients given RIST vs 49 [82%] of 60 patients given control), thrombocytopenia (45 [67%] vs 41 [68%]), and anaemia (39 [58%] vs 38 [63%]). Nine serious treatment-related adverse events were reported (five patients given control and four patients given RIST). There were no treatment-related deaths in the control group and one in the RIST group (multiorgan failure). INTERPRETATION: RIST-rNB-2011 demonstrated that targeting of MYCN-amplified relapsed or refractory neuroblastoma with a pathway-directed metronomic combination of a multkinase inhibitor and an mTOR inhibitor can improve progression-free survival and overall survival. This exclusive efficacy in MYCN-amplified, relapsed neuroblastoma warrants further investigation in the first-line setting. FUNDING: Deutsche Krebshilfe.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Dasatinib , Irinotécan , Récidive tumorale locale , Neuroblastome , Sirolimus , Témozolomide , Humains , Témozolomide/administration et posologie , Témozolomide/usage thérapeutique , Irinotécan/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Mâle , Femelle , Neuroblastome/traitement médicamenteux , Neuroblastome/mortalité , Neuroblastome/anatomopathologie , Neuroblastome/génétique , Enfant d'âge préscolaire , Enfant , Dasatinib/administration et posologie , Dasatinib/usage thérapeutique , Dasatinib/effets indésirables , Adolescent , Récidive tumorale locale/traitement médicamenteux , Récidive tumorale locale/anatomopathologie , Nourrisson , Adulte , Sirolimus/administration et posologie , Sirolimus/usage thérapeutique , Jeune adulte , Allemagne , Résistance aux médicaments antinéoplasiques , Survie sans progression
18.
J Cancer Res Clin Oncol ; 150(5): 271, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38780840

RÉSUMÉ

OBJECTIVE: Previous research has shown that both temozolomide (TMZ) and PD-1/L1 inhibitors (PD-1/L1) alone exhibit certain potential in the treatment of non-small cell lung cancer (NSCLC) with brain metastases (BM), in this study, we will explore combining the two in order to seek new effective treatment options for NSCLC with BM. MATERIAL AND METHODS: During 2021.1 to 2023.12, we collected the date of these pretreated-NSCLC with BM who accept the treatment of TMZ and PD-1/L1, the objective response ratio (ORR), progression-free survival (PFS) and overall survival (OS) were set as the primary endpoint, meanwhile, the toxicity of such regimen was also recorded. RESULTS: About 42 patients are enrolled, our primary analysis demonstrated that the ORR of such regimen toward NSCLC with BM was 26.19%, with Approximate intracranial and extracranial lesion ORR was 6% and 20% respectively, the DCR was about 64.29%, the mean PFS and OS was about 4 m and 8.5 m. Further analysis indicated that the efficiency correlated with the diagnosis-Specific Graded Prognostic Assessment (ds-GPA) score. Moreover, the toxicity can also be tolerated, indicating the application potential of such regimen against NSCLC with BM. CONCLUSIONS: Our results exhibited that with tolerated toxicity, the combination of TMZ and PD-1/L1 shows promising efficiency against NSCLC with BM, this would be of great significance for the treatment of NSCLC with brain metastasis. However, due to the limitation of sample and retrospective property, the real value of such regimen needed to be further confirmed in the future.


Sujet(s)
Tumeurs du cerveau , Carcinome pulmonaire non à petites cellules , Inhibiteurs de points de contrôle immunitaires , Tumeurs du poumon , Témozolomide , Humains , Témozolomide/usage thérapeutique , Témozolomide/administration et posologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Tumeurs du cerveau/secondaire , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Études rétrospectives , Mâle , Adulte d'âge moyen , Femelle , Sujet âgé , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/effets indésirables , Inhibiteurs de points de contrôle immunitaires/administration et posologie , Adulte , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Antigène CD274/antagonistes et inhibiteurs
19.
Cell Death Dis ; 15(5): 366, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38806469

RÉSUMÉ

Glioblastoma (GBM) is the most aggressive and lethal brain tumor in adults. This study aimed to investigate the functional significance of aryl hydrocarbon receptor nuclear translocator (ARNT) in the pathogenesis of GBM. Analysis of public datasets revealed ARNT is upregulated in GBM tissues compared to lower grade gliomas or normal brain tissues. Higher ARNT expression correlated with the mesenchymal subtype and poorer survival in GBM patients. Silencing ARNT using lentiviral shRNAs attenuated the proliferative, invasive, and stem-like capabilities of GBM cell lines, while ARNT overexpression enhanced these malignant phenotypes. Single-cell RNA sequencing uncovered that ARNT is highly expressed in a stem-like subpopulation and is involved in regulating glycolysis, hypoxia response, and stress pathways. Mechanistic studies found ARNT activates p38 mitogen-activated protein kinase (MAPK) signaling to promote chemoresistance in GBM cells. Disrupting the ARNT/p38α protein interaction via the ARNT PAS-A domain restored temozolomide sensitivity. Overall, this study demonstrates ARNT functions as an oncogenic driver in GBM pathogenesis and represents a promising therapeutic target.


Sujet(s)
Translocateur nucléaire du récepteur des hydrocarbures aromatiques , Résistance aux médicaments antinéoplasiques , Glioblastome , Humains , Glioblastome/traitement médicamenteux , Glioblastome/génétique , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Translocateur nucléaire du récepteur des hydrocarbures aromatiques/métabolisme , Translocateur nucléaire du récepteur des hydrocarbures aromatiques/génétique , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Animaux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mitogen-Activated Protein Kinase 14/métabolisme , Mitogen-Activated Protein Kinase 14/génétique , Souris , Régulation de l'expression des gènes tumoraux , Témozolomide/pharmacologie , Témozolomide/usage thérapeutique , Souris nude , Transduction du signal/effets des médicaments et des substances chimiques
20.
Eur J Pharm Biopharm ; 200: 114326, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38759897

RÉSUMÉ

Glioblastoma (GBM) is a highly deadly brain tumor that does not respond satisfactorily to conventional treatment. The non-alkylating agent gemcitabine (GEM) has been proposed for treating GBM. It can overcome MGMT protein-mediated resistance, a major limitation of conventional therapy with the alkylating agent temozolomide (TMZ). However, GEM's high systemic toxicity and poor permeability across the blood-brain barrier (BBB) pose significant challenges for its delivery to the brain. Thus, mucoadhesive poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) coated with chitosan (CH), suitable for intranasal GEM delivery, were proposed in this work. A central composite design (CCD) was implemented for NPs optimization, and NPs with appropriate characteristics for intranasal administration were obtained. in vitro studies revealed that the NPs possess excellent mucoadhesive properties and the ability to selectively release GEM in the simulated tumor tissue environment. in vitro studies using two human GBM cell lines (U215 and T98G) revealed the NPs' ability to promote GEM's antiproliferative activity to sensitize cells to the effect of TMZ. The findings of this work demonstrate that the developed CH-GEM-NPs are suitable delivery systems for GEM, both as a single therapy and as a chemosensitizer to the GBM gold standard therapy.


Sujet(s)
Tumeurs du cerveau , Chitosane , Désoxycytidine , Repositionnement des médicaments , , Glioblastome , Nanoparticules , Copolymère d'acide poly(lactique-co-glycolique) , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Désoxycytidine/analogues et dérivés , Désoxycytidine/administration et posologie , Désoxycytidine/pharmacologie , Désoxycytidine/composition chimique , Humains , Chitosane/composition chimique , Copolymère d'acide poly(lactique-co-glycolique)/composition chimique , Nanoparticules/composition chimique , Lignée cellulaire tumorale , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/métabolisme , Repositionnement des médicaments/méthodes , Témozolomide/administration et posologie , Témozolomide/pharmacologie , Témozolomide/pharmacocinétique , Administration par voie nasale , Antimétabolites antinéoplasiques/administration et posologie , Antimétabolites antinéoplasiques/pharmacologie , Antimétabolites antinéoplasiques/composition chimique , Antimétabolites antinéoplasiques/pharmacocinétique , Vecteurs de médicaments/composition chimique , Barrière hémato-encéphalique/métabolisme , Libération de médicament
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...