Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 16.498
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.
Cells ; 13(13)2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-38995006

RÉSUMÉ

Immunotherapies have shown significant promise as an impactful strategy in cancer treatment. However, in glioblastoma multiforme (GBM), the most prevalent primary brain tumor in adults, these therapies have demonstrated lower efficacy than initially anticipated. Consequently, there is an urgent need for strategies to enhance the effectiveness of immune treatments. AURKA has been identified as a potential drug target for GBM treatment. An analysis of the GBM cell transcriptome following AURKA inhibition revealed a potential influence on the immune system. Our research revealed that AURKA influenced PD-L1 levels in various GBM model systems in vitro and in vivo. Disrupting AURKA function genetically led to reduced PD-L1 levels and increased MHC-I expression in both established and patient-derived xenograft GBM cultures. This process involved both transcriptional and non-transcriptional pathways, partly implicating GSK3ß. Interfering with AURKA also enhanced NK-cell-mediated elimination of GBM by reducing PD-L1 expression, as evidenced in rescue experiments. Furthermore, using a mouse model that mimics GBM with patient-derived cells demonstrated that Alisertib decreased PD-L1 expression in living organisms. Combination therapy involving anti-PD-1 treatment and Alisertib significantly prolonged overall survival compared to vehicle treatment. These findings suggest that targeting AURKA could have therapeutic implications for modulating the immune environment within GBM cells.


Sujet(s)
Aurora kinase A , Antigène CD274 , Glioblastome , Cellules tueuses naturelles , Aurora kinase A/métabolisme , Aurora kinase A/antagonistes et inhibiteurs , Humains , Glioblastome/anatomopathologie , Glioblastome/traitement médicamenteux , Glioblastome/immunologie , Glioblastome/génétique , Antigène CD274/métabolisme , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/effets des médicaments et des substances chimiques , Cellules tueuses naturelles/métabolisme , Animaux , Souris , Lignée cellulaire tumorale , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Azépines/pharmacologie , Pyrimidines/pharmacologie , Cytotoxicité immunologique/effets des médicaments et des substances chimiques , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
3.
Int J Nanomedicine ; 19: 6757-6776, 2024.
Article de Anglais | MEDLINE | ID: mdl-38983132

RÉSUMÉ

Glioma is a primary malignant tumor in the central nervous system. In recent years, the treatment of glioma has developed rapidly, but the overall survival of glioma patients has not significantly improved. Due to the presence of the blood-brain barrier and intracranial tumor barrier, many drugs with good effects to cure glioma in vitro cannot be accurately transported to the corresponding lesions. In order to enable anti-tumor drugs to overcome the barriers and target glioma, nanodrug delivery systems have emerged recently. It is gratifying that liposomes, as a multifunctional nanodrug delivery carrier, which can be compatible with hydrophilic and hydrophobic drugs, easily functionalized by various targeted ligands, biodegradable, and hypoimmunogenic in vivo, has become a quality choice to solve the intractable problem of glioma medication. Therefore, we focused on the liposome nanodrug delivery system, and summarized its current research progress in glioma. Hopefully, this review may provide new ideas for the research and development of liposome-based nanomaterials for the clinical treatment of glioma.


Sujet(s)
Antinéoplasiques , Barrière hémato-encéphalique , Tumeurs du cerveau , Gliome , Liposomes , Nanostructures , Gliome/traitement médicamenteux , Liposomes/composition chimique , Humains , Tumeurs du cerveau/traitement médicamenteux , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Antinéoplasiques/composition chimique , Antinéoplasiques/administration et posologie , Animaux , Nanostructures/composition chimique , Nanostructures/usage thérapeutique , Systèmes de délivrance de médicaments/méthodes , Nanomédecine/méthodes , Vecteurs de médicaments/composition chimique
4.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(6): 1088-1097, 2024 Jun 20.
Article de Chinois | MEDLINE | ID: mdl-38977338

RÉSUMÉ

OBJECTIVE: To investigate the effect of the aqueous extract of Chuan Xiong Rhizoma (CR) on brain metastasis of melanoma B16F10 cells in mice. METHODS: C57BL/6J mouse models of brain metastasis of melanoma were established by ultrasound-guided intraventricular injection of Luc-labeled B16F10 cells, and brain tumor growth was monitored by in vivo imaging. The mouse models were then randomized for daily gavage of saline or aqueous extract of CR (equivalent crude drug concentration of 1 mg/g). Behavioral tests were used to evaluate the neuroprotective effects of CR in the tumor-bearing mice, and the changes in proteins associated with blood-brain barrier integrity, neuronal cell proliferation and apoptosis, and microglial cell apoptosis and activation were observed using immunofluorescence assay. The efficacy of CR combined with temozolomide (25 mg/kg) against brain metastases of B16F10 cells was observed by in vivo imaging. RESULTS: CR-treated mouse models did not show obvious progression of brain metastases and had a reduced rate of body weight loss and lowered protein expressions of ZO-1, claudin-5, occludin, P-gp, TNF-α, AQP4 and PDGFRß. In the behavioral tests, the CR-treated mice showed prolonged stay on the wooden stick with a shortened time of sticky stick removal. Immunofluorescence assay showed increased proliferation and decreased apoptosis of neuronal cells and microglia in CR-treated mice. CR treatment significantly increased the levels of CD86, CD206, IL-4 and IL-10 and decreased the levels of CD163 and IL-1ß in the microenvironment of brain metastases. The mice receiving combined treatments with CR and temozolomide showed significantly lower intensity of fluorescent signals in the brain than those treated with temozolomide alone. CONCLUSION: CR does not promote brain metastasis of melanoma while inducing opening of the blood-brain barrier, and its combined use with TMZ results in enhanced inhibition against brain metastasis of melanoma B16F10 cells in mice.


Sujet(s)
Tumeurs du cerveau , Médicaments issus de plantes chinoises , Souris de lignée C57BL , Témozolomide , Animaux , Témozolomide/pharmacologie , Souris , Tumeurs du cerveau/secondaire , Tumeurs du cerveau/traitement médicamenteux , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/usage thérapeutique , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Dacarbazine/analogues et dérivés , Dacarbazine/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mélanome expérimental/traitement médicamenteux , Mélanome expérimental/anatomopathologie
5.
J Transl Med ; 22(1): 648, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38987805

RÉSUMÉ

Glioma is the most common malignant tumor in central nervous system, with significant health burdens to patients. Due to the intrinsic characteristics of glioma and the lack of breakthroughs in treatment modalities, the prognosis for most patients remains poor. This results in a heavy psychological and financial load worldwide. In recent years, cannabidiol (CBD) has garnered widespread attention and research due to its anti-tumoral, anti-inflammatory, and neuroprotective properties. This review comprehensively summarizes the preclinical and clinical research on the use of CBD in glioma therapy, as well as the current status of nanomedicine formulations of CBD, and discusses the potential and challenges of CBD in glioma therapy in the future.


Sujet(s)
Cannabidiol , Gliome , Cannabidiol/usage thérapeutique , Cannabidiol/pharmacologie , Humains , Gliome/traitement médicamenteux , Gliome/anatomopathologie , Animaux , , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Nanomédecine/méthodes
6.
BMJ Open ; 14(7): e078335, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38969367

RÉSUMÉ

BACKGROUND: Patients with advanced non-small-cell lung cancer (NSCLC) with activating mutations in the epidermal growth factor receptor (EGFR) gene are a heterogeneous population who often develop brain metastases (BM). The optimal management of patients with asymptomatic brain metastases is unclear given the activity of newer-generation targeted therapies in the central nervous system. We present a protocol for an individual patient data (IPD) prospective meta-analysis to evaluate whether the addition of stereotactic radiosurgery (SRS) before osimertinib treatment will lead to better control of intracranial metastatic disease. This is a clinically relevant question that will inform practice. METHODS: Randomised controlled trials will be eligible if they include participants with BM arising from EGFR-mutant NSCLC and suitable to receive osimertinib both in the first-line and second-line settings (P); comparisons of SRS followed by osimertinib versus osimertinib alone (I, C) and intracranial disease control included as an endpoint (O). Systematic searches of Medline (Ovid), Embase (Ovid), Cochrane Central Register of Controlled Trials (CENTRAL), CINAHL (EBSCO), PsychInfo, ClinicalTrials.gov and the WHO's International Clinical Trials Registry Platform's Search Portal will be undertaken. An IPD meta-analysis will be performed using methodologies recommended by the Cochrane Collaboration. The primary outcome is intracranial progression-free survival, as determined by response assessment in neuro-oncology-BM criteria. Secondary outcomes include overall survival, time to whole brain radiotherapy, quality of life, and adverse events of special interest. Effect differences will be explored among prespecified subgroups. ETHICS AND DISSEMINATION: Approved by each trial's ethics committee. Results will be relevant to clinicians, researchers, policymakers and patients, and will be disseminated via publications, presentations and media releases. PROSPERO REGISTRATION: CRD42022330532.


Sujet(s)
Acrylamides , Dérivés de l'aniline , Tumeurs du cerveau , Carcinome pulmonaire non à petites cellules , Récepteurs ErbB , Tumeurs du poumon , Radiochirurgie , Revues systématiques comme sujet , Humains , Acrylamides/usage thérapeutique , Dérivés de l'aniline/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Tumeurs du cerveau/secondaire , Tumeurs du cerveau/génétique , Tumeurs du cerveau/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/secondaire , Association thérapeutique , Récepteurs ErbB/génétique , Indoles , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Méta-analyse comme sujet , Mutation , Études prospectives , Pyrimidines , Radiochirurgie/méthodes , Essais contrôlés randomisés comme sujet , Plan de recherche
7.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-39000069

RÉSUMÉ

Lung cancer is a leading cause of cancer-related morbidity and mortality worldwide. Metastases in the brain are a common hallmark of advanced stages of the disease, contributing to a dismal prognosis. Lung cancer can be broadly classified as either small cell lung cancer (SCLC) or non-small cell lung cancer (NSCLC). NSCLC represents the most predominant histology subtype of lung cancer, accounting for the majority of lung cancer cases. Recent advances in molecular genetics, coupled with innovations in small molecule drug discovery strategies, have facilitated both the molecular classification and precision targeting of NSCLC based on oncogenic driver mutations. Furthermore, these precision-based strategies have demonstrable efficacy across the blood-brain barrier, leading to positive outcomes in patients with brain metastases. This review provides an overview of the clinical features of lung cancer brain metastases, as well as the molecular mechanisms that drive NSCLC oncogenesis. We also explore how precision medicine-based strategies can be leveraged to improve NSCLC brain metastases.


Sujet(s)
Tumeurs du cerveau , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du cerveau/secondaire , Tumeurs du cerveau/génétique , Tumeurs du cerveau/traitement médicamenteux , Médecine de précision/méthodes , Mutation , Barrière hémato-encéphalique/métabolisme , Antinéoplasiques/usage thérapeutique
8.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-39000148

RÉSUMÉ

The metabolism of glioma cells exhibits significant heterogeneity and is partially responsible for treatment outcomes. Given this variability, we hypothesized that the effectiveness of treatments targeting various metabolic pathways depends on the bioenergetic profiles and mitochondrial status of glioma cells. To this end, we analyzed mitochondrial biomass, mitochondrial protein density, oxidative phosphorylation (OXPHOS), and glycolysis in a panel of eight glioma cell lines. Our findings revealed considerable variability: mitochondrial biomass varied by up to 3.2-fold, the density of mitochondrial proteins by up to 2.1-fold, and OXPHOS levels by up to 7.3-fold across the cell lines. Subsequently, we stratified glioma cell lines based on their mitochondrial status, OXPHOS, and bioenergetic fitness. Following this stratification, we utilized 16 compounds targeting key bioenergetic, mitochondrial, and related pathways to analyze the associations between induced changes in cell numbers, proliferation, and apoptosis with respect to their steady-state mitochondrial and bioenergetic metrics. Remarkably, a significant fraction of the treatments showed strong correlations with mitochondrial biomass and the density of mitochondrial proteins, suggesting that mitochondrial status may reflect glioma cell sensitivity to specific treatments. Overall, our results indicate that mitochondrial status and bioenergetics are linked to the efficacy of treatments targeting metabolic pathways in glioma.


Sujet(s)
Biomasse , Métabolisme énergétique , Gliome , Mitochondries , Protéines mitochondriales , Phosphorylation oxydative , Gliome/métabolisme , Gliome/anatomopathologie , Humains , Lignée cellulaire tumorale , Mitochondries/métabolisme , Protéines mitochondriales/métabolisme , Prolifération cellulaire , Glycolyse , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/traitement médicamenteux , Apoptose
9.
Acta Neuropathol Commun ; 12(1): 118, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-39014476

RÉSUMÉ

Background Neurotrophic tropomyosin receptor kinase (NTRK) gene fusions are found in 1% of gliomas across children and adults. TRK inhibitors are promising therapeutic agents for NTRK-fused gliomas because they are tissue agnostic and cross the blood-brain barrier (BBB). Methods We investigated twelve NGS-verified NTRK-fused gliomas from a single institute, Seoul National University Hospital. Results The patient cohort included six children (aged 1-15 years) and six adults (aged 27-72 years). NTRK2 fusions were found in ten cerebral diffuse low-grade and high-grade gliomas (DLGGs and DHGGs, respectively), and NTRK1 fusions were found in one cerebral desmoplastic infantile ganglioglioma and one spinal DHGG. In this series, the fusion partners of NTRK2 were HOOK3, KIF5A, GKAP1, LHFPL3, SLMAP, ZBTB43, SPECC1L, FKBP15, KANK1, and BCR, while the NTRK1 fusion partners were TPR and TPM3. DLGGs tended to harbour only an NTRK fusion, while DHGGs exhibited further genetic alterations, such as TERT promoter/TP53/PTEN mutation, CDKN2A/2B homozygous deletion, PDGFRA/KIT/MDM4/AKT3 amplification, or multiple chromosomal copy number aberrations. Four patients received adjuvant TRK inhibitor therapy (larotrectinib, repotrectinib, or entrectinib), among which three also received chemotherapy (n = 2) or proton therapy (n = 1). The treatment outcomes for patients receiving TRK inhibitors varied: one child who received larotrectinib for residual DLGG maintained stable disease. In contrast, another child with DHGG in the spinal cord experienced multiple instances of tumour recurrence. Despite treatment with larotrectinib, ultimately, the child died as a result of tumour progression. An adult patient with glioblastoma (GBM) treated with entrectinib also experienced tumour progression and eventually died. However, there was a successful outcome for a paediatric patient with DHGG who, after a second gross total tumour removal followed by repotrectinib treatment, showed no evidence of disease. This patient had previously experienced relapse after the initial surgery and underwent autologous peripheral blood stem cell therapy with carboplatin/thiotepa and proton therapy. Conclusions Our study clarifies the distinct differences in the pathology and TRK inhibitor response between LGG and HGG with NTRK fusions.


Sujet(s)
Inhibiteurs de protéines kinases , Pyrazoles , Récepteur trkB , Humains , Mâle , Femelle , Enfant , Enfant d'âge préscolaire , Adulte , Adolescent , Adulte d'âge moyen , Sujet âgé , Nourrisson , Récepteur trkB/génétique , Récepteur trkB/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Pyrazoles/usage thérapeutique , Récepteur trkA/génétique , Récepteur trkA/antagonistes et inhibiteurs , Gliome/génétique , Gliome/anatomopathologie , Gliome/traitement médicamenteux , Pyrimidines/usage thérapeutique , Protéines de fusion oncogènes/génétique , Benzamides/usage thérapeutique , Glycoprotéines membranaires/génétique , Tumeurs du système nerveux central/génétique , Tumeurs du système nerveux central/traitement médicamenteux , Tumeurs du système nerveux central/anatomopathologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Indazoles
10.
CNS Neurosci Ther ; 30(7): e14839, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39021040

RÉSUMÉ

BACKGROUND: The role of the unfolded protein response (UPR) has been progressively unveiled over the last decade and several studies have investigated its implication in glioblastoma (GB) development. The UPR restores cellular homeostasis by triggering the folding and clearance of accumulated misfolded proteins in the ER consecutive to endoplasmic reticulum stress. In case it is overwhelmed, it induces apoptotic cell death. Thus, holding a critical role in cell fate decisions. METHODS: This article, reviews how the UPR is implicated in cell homeostasis maintenance, then surveils the evidence supporting the UPR involvement in GB genesis, progression, angiogenesis, GB stem cell biology, tumor microenvironment modulation, extracellular matrix remodeling, cell fate decision, invasiveness, and grading. Next, it concurs the evidence showing how the UPR mediates GB chemoresistance-related mechanisms. RESULTS: The UPR stress sensors IRE1, PERK, and ATF6 with their regulator GRP78 are upregulated in GB compared to lower grade gliomas and normal brain tissue. They are activated in response to oncogenes and are implicated at different stages of GB progression, from its genesis to chemoresistance and relapse. The UPR arms can be effectors of apoptosis as mediators or targets. CONCLUSION: Recent research has established the role of the UPR in GB pathophysiology and chemoresistance. Targeting its different sensors have shown promising in overcoming GB chomo- and radioresistance and inducing apoptosis.


Sujet(s)
Tumeurs du cerveau , Résistance aux médicaments antinéoplasiques , Chaperonne BiP du réticulum endoplasmique , Glioblastome , Réponse aux protéines mal repliées , Humains , Réponse aux protéines mal repliées/effets des médicaments et des substances chimiques , Réponse aux protéines mal repliées/physiologie , Glioblastome/traitement médicamenteux , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Résistance aux médicaments antinéoplasiques/physiologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Animaux , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique
11.
J Transl Med ; 22(1): 667, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39026284

RÉSUMÉ

BACKGROUND: In the fight against GBM, drug repurposing emerges as a viable and time-saving approach to explore new treatment options. Chlorpromazine, an old antipsychotic medication, has recently arisen as a promising candidate for repositioning in GBM therapy in addition to temozolomide, the first-line standard of care. We previously demonstrated the antitumor efficacy of chlorpromazine and its synergistic effects with temozolomide in suppressing GBM cell malignant features in vitro. This prompted us to accomplish a Phase II clinical trial to evaluate the efficacy and safety of adding chlorpromazine to temozolomide in GBM patients with unmethylated MGMT gene promoter. In this in vitro study, we investigate the potential role of chlorpromazine in overcoming temozolomide resistance. METHODS: In our experimental set, we analyzed Connexin-43 expression at both the transcriptional and protein levels in control- and chlorpromazine-treated GBM cells. DNA damage and subsequent repair were assessed by immunofluorescence of γ-H2AX and Reverse-Phase Protein microArrays in chlorpromazine treated GBM cell lines. To elucidate the relationship between DNA repair systems and chemoresistance, we analyzed a signature of DNA repair genes in GBM cells after treatment with chlorpromazine, temozolomide and Connexin-43 downregulation. RESULTS: Chlorpromazine treatment significantly downregulated connexin-43 expression in GBM cells, consequently compromising connexin-dependent cellular resilience, and ultimately contributing to cell death. In line with this, we observed concordant post-translational modifications of molecular determinants involved in DNA damage and repair pathways. Our evaluation of DNA repair genes revealed that temozolomide elicited an increase, while chlorpromazine, as well as connexin-43 silencing, a decrease in DNA repair gene expression in GBM cells. CONCLUSIONS: Chlorpromazine potentiates the cytotoxic effects of the alkylating agent temozolomide through a mechanism involving downregulation of Cx43 expression and disruption of the cell cycle arrest essential for DNA repair processes. This finding suggests that chlorpromazine may be a potential therapeutic strategy to overcome TMZ resistance in GBM cells by inhibiting their DNA repair mechanisms.


Sujet(s)
Chlorpromazine , Connexine 43 , Réparation de l'ADN , Résistance aux médicaments antinéoplasiques , Glioblastome , Témozolomide , Chlorpromazine/pharmacologie , Chlorpromazine/usage thérapeutique , Humains , Témozolomide/pharmacologie , Témozolomide/usage thérapeutique , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Glioblastome/génétique , Réparation de l'ADN/effets des médicaments et des substances chimiques , Connexine 43/métabolisme , Connexine 43/génétique , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Synergie des médicaments , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/génétique
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.
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
15.
Int J Mol Sci ; 25(13)2024 Jun 22.
Article de Anglais | MEDLINE | ID: mdl-38999983

RÉSUMÉ

The synthesis, biochemical evaluation and radiosynthesis of a cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitor and radioligand was performed. NT431, a newly synthesized 4-fluorobenzyl-abemaciclib, exhibited high potency to CDK4/6 and against four cancer cell lines with IC50 similar to that of the parent abemaciclib. We performed a two-step one-pot radiosynthesis to produce [18F]NT431 with good radiochemical yield (9.6 ± 3%, n = 3, decay uncorrected), high radiochemical purity (>95%), and high molar activity (>370 GBq/µmol (>10.0 Ci/µmol). In vitro autoradiography confirmed the specific binding of [18F]NT431 to CDK4/6 in brain tissues. Dynamic PET imaging supports that both [18F]NT431 and the parent abemaciclib crossed the BBB albeit with modest brain uptake. Therefore, we conclude that it is unlikely that NT431 or abemaciclib (FDA approved drug) can accumulate in the brain in sufficient concentrations to be potentially effective against breast cancer brain metastases or brain cancers. However, despite the modest BBB penetration, [18F]NT431 represents an important step towards the development and evaluation of a new generation of CDK4/6 inhibitors with superior BBB penetration for the treatment and visualization of CDK4/6 positive tumors in the CNS. Also, [18F]NT431 may have potential application in peripheral tumors such as breast cancer and other CDK4/6 positive tumors.


Sujet(s)
Aminopyridines , Benzimidazoles , Tumeurs du cerveau , Kinase-4 cycline-dépendante , Kinase-6 cycline-dépendante , Tomographie par émission de positons , Inhibiteurs de protéines kinases , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Kinase-4 cycline-dépendante/métabolisme , Humains , Tomographie par émission de positons/méthodes , Tumeurs du cerveau/imagerie diagnostique , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/enzymologie , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/métabolisme , Lignée cellulaire tumorale , Benzimidazoles/pharmacologie , Benzimidazoles/composition chimique , Aminopyridines/composition chimique , Aminopyridines/pharmacologie , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/synthèse chimique , Animaux , Radiopharmaceutiques/composition chimique , Radio-isotopes du fluor/composition chimique , Encéphale/imagerie diagnostique , Encéphale/métabolisme , Souris , Femelle
16.
ACS Appl Mater Interfaces ; 16(28): 35925-35935, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38950334

RÉSUMÉ

The development of efficient theranostic nanoagents for the precise diagnosis and targeted therapy of glioblastoma (GBM) remains a big challenge. Herein, we designed and developed porphyrin-based organic nanoparticles (PNP NPs) with strong emission in the near-infrared IIa window (NIR-IIa) for orthotopic GBM theranostics. PNP NPs possess favorable photoacoustic and photothermal properties, high photostability, and low toxicity. After modification with the RGD peptide, the obtained PNPD NPs exhibited enhanced blood-brain barrier (BBB) penetration capability and GBM targeting ability. NIR-IIa imaging was employed to monitor the in vivo biodistribution and accumulation of the nanoparticles, revealing a significant enhancement in penetration depth and signal-to-noise ratio. Both in vitro and in vivo results demonstrated that PNPD NPs effectively inhibited the proliferation of tumor cells and induced negligible side effects in normal brain tissues. In general, the work presented a kind of brain-targeted porphyrin-based NPs with NIR-IIa fluorescence for orthotopic glioblastoma theranostics, showing promising prospects for clinical translation.


Sujet(s)
Glioblastome , Nanoparticules , Porphyrines , Nanomédecine théranostique , Glioblastome/imagerie diagnostique , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Glioblastome/métabolisme , Animaux , Nanoparticules/composition chimique , Humains , Porphyrines/composition chimique , Porphyrines/pharmacologie , Souris , Tumeurs du cerveau/imagerie diagnostique , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Lignée cellulaire tumorale , Rayons infrarouges , Distribution tissulaire , Barrière hémato-encéphalique/métabolisme , Souris nude , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Fluorescence
17.
Cell Rep Med ; 5(7): 101630, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38955178

RÉSUMÉ

Recurrent high-grade gliomas (rHGGs) have a dismal prognosis, where the maximum tolerated dose (MTD) of IV terameprocol (5 days/month), a transcriptional inhibitor of specificity protein 1 (Sp1)-regulated proteins, is 1,700 mg/day with median area under the plasma concentration-time curve (AUC) of 31.3 µg∗h/mL. Given potentially increased efficacy with sustained systemic exposure and challenging logistics of daily IV therapy, here we investigate oral terameprocol for rHGGs in a multicenter, phase 1 trial (GATOR). Using a 3 + 3 dose-escalation design, we enroll 20 patients, with median age 60 years (range 31-80), 70% male, and median one relapse (range 1-3). Fasting patients tolerate 1,200 mg/day (n = 3), 2,400 mg/day (n = 6), 3,600 mg/day (n = 3), and 6,000 mg/day (n = 2) oral doses without major toxicities. However, increased dosage does not lead to increased systemic exposure, including in fed state (6,000 mg/day, n = 4), with maximal AUC <5 µg∗h/mL. These findings warrant trials investigating approaches that provide sustained systemic levels of transcription inhibitors to exploit their therapeutic potential. This study was registered at ClinicalTrials.gov (NCT02575794).


Sujet(s)
Tumeurs du cerveau , Gliome , Humains , Mâle , Adulte d'âge moyen , Gliome/traitement médicamenteux , Gliome/anatomopathologie , Adulte , Femelle , Sujet âgé , Administration par voie orale , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Sujet âgé de 80 ans ou plus , Récidive tumorale locale/traitement médicamenteux , Récidive tumorale locale/anatomopathologie , Grading des tumeurs , Dose maximale tolérée
18.
Int J Oncol ; 65(2)2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38994761

RÉSUMÉ

Glioblastoma (GBM) is the most common malignancy of the central nervous system in adults. The current standard of care includes surgery, radiation therapy, temozolomide; and tumor­treating fields leads to dismal overall survival. There are far limited treatments upon recurrence. Therapies to date are ineffective as a result of several factors, including the presence of the blood­brain barrier, blood tumor barrier, glioma stem­like cells and genetic heterogeneity in GBM. In the present review, the potential mechanisms that lead to treatment resistance in GBM and the measures which have been taken so far to attempt to overcome the resistance were discussed. The complex biology of GBM and lack of comprehensive understanding of the development of therapeutic resistance in GBM demands discovery of novel antigens that are targetable and provide effective therapeutic strategies.


Sujet(s)
Barrière hémato-encéphalique , Tumeurs du cerveau , Résistance aux médicaments antinéoplasiques , Glioblastome , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Glioblastome/génétique , Humains , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/thérapie , Barrière hémato-encéphalique/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/anatomopathologie , Thérapie moléculaire ciblée/méthodes
19.
JCO Precis Oncol ; 8: e2300590, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38976815

RÉSUMÉ

PURPOSE: Midline low-grade gliomas (mLGGs) of early childhood have a poorer prognosis compared with tumors of other localizations and in older patients. LGGs are associated with aberrant activation of RAS-RAF-MEK pathway, and pharmacological inhibition of the pathway has therapeutic promise. The aim of this study was clinical and molecular characterization of infantile mLGGs, with emphasis on the efficacy of targeted kinase inhibition. PATIENTS AND METHODS: This study enrolled 40 patients with mLGG age <3 years. The majority of the patients (30/40) received first-line chemotherapy (CT) as per International Society of Paediatric Oncology LGG 2004 guidelines. In all patients, molecular genetic investigation of tumor tissue by polymerase chain reaction and RNA sequencing was performed. The median follow-up was 3.5 years. RESULTS: First-line CT failed in 24 of 30 recipients. The identified molecular profiles included KIAA1549::BRAF fusions in 26 patients, BRAF V600E in six patients, FGFR1::TACC1 fusions in two patients, and rare fusion transcripts in four patients. At disease progression, targeted therapy (TT) was initiated in 27 patients (22 patients received trametinib) on the basis of molecular findings. TT was administered for a median of 16 months, with partial response achieved in 12 of 26 (46%) patients in which response was evaluated. Severe adverse events were detected only on trametinib monotherapy: acute damage of GI or urinary mucosa complicated by hemorrhage and development of transfusion-dependent anemia in four patients and grade 3 skin toxicity in three patients. CONCLUSION: mLGGs of early childhood are often aggressive tumors, resistant to CT, and frequently require alternative treatment. The majority of patients harbor druggable molecular targets and respond to molecular TT.


Sujet(s)
Tumeurs du cerveau , Gliome , Thérapie moléculaire ciblée , Humains , Gliome/génétique , Gliome/traitement médicamenteux , Mâle , Femelle , Nourrisson , Enfant d'âge préscolaire , Thérapie moléculaire ciblée/méthodes , Tumeurs du cerveau/génétique , Tumeurs du cerveau/traitement médicamenteux
20.
Int J Nanomedicine ; 19: 6999-7014, 2024.
Article de Anglais | MEDLINE | ID: mdl-39011386

RÉSUMÉ

Introduction: Glioblastoma multiforme (GBM), a highly invasive and prognostically challenging brain cancer, poses a significant hurdle for current treatments due to the existence of the blood-brain barrier (BBB) and the difficulty to maintain an effective drug accumulation in deep GBM lesions. Methods: We present a biomimetic nanoplatform with angiopep-2-modified macrophage membrane, loaded with indocyanine green (ICG) templated self-assembly of SN38 (AM-NP), facilitating active tumor targeting and effective blood-brain barrier penetration through specific ligand-receptor interaction. Results: Upon accumulation at tumor sites, these nanoparticles achieved high drug concentrations. Subsequent combination of laser irradiation and release of chemotherapy agent SN38 induced a synergistic chemo-photothermal therapy. Compared to bare nanoparticles (NPs) lacking cell membrane encapsulation, AM-NPs significantly suppressed tumor growth, markedly enhanced survival rates, and exhibited excellent biocompatibility with minimal side effects. Conclusion: This NIR-activatable biomimetic camouflaging macrophage membrane-based nanoparticles enhanced drug delivery targeting ability through modifications of macrophage membranes and specific ligands. It simultaneously achieved synergistic chemo-photothermal therapy, enhancing treatment effectiveness. Compared to traditional treatment modalities, it provided a precise, efficient, and synergistic method that might have contributed to advancements in glioblastoma therapy.


Sujet(s)
Barrière hémato-encéphalique , Tumeurs du cerveau , Libération de médicament , Glioblastome , Vert indocyanine , Nanoparticules , Thérapie photothermique , Glioblastome/thérapie , Glioblastome/traitement médicamenteux , Glioblastome/métabolisme , Animaux , Vert indocyanine/composition chimique , Vert indocyanine/pharmacocinétique , Vert indocyanine/pharmacologie , Tumeurs du cerveau/thérapie , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/métabolisme , Humains , Lignée cellulaire tumorale , Souris , Nanoparticules/composition chimique , Thérapie photothermique/méthodes , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Irinotécan/pharmacocinétique , Irinotécan/composition chimique , Irinotécan/pharmacologie , Peptides/composition chimique , Peptides/pharmacologie , Peptides/pharmacocinétique , Rayons infrarouges , Matériaux biomimétiques/composition chimique , Matériaux biomimétiques/pharmacocinétique , Matériaux biomimétiques/pharmacologie , Systèmes de délivrance de médicaments/méthodes , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Souris nude , Association thérapeutique/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE