Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42.285
Filter
1.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article in English | MEDLINE | ID: mdl-39000485

ABSTRACT

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.


Subject(s)
Glioblastoma , Lipoproteins, HDL , Nanoparticles , Spheroids, Cellular , Xanthones , Humans , Xanthones/chemistry , Xanthones/pharmacology , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/metabolism , Cell Line, Tumor , Nanoparticles/chemistry , Lipoproteins, HDL/chemistry , Lipoproteins, HDL/metabolism , Spheroids, Cellular/drug effects , Drug Carriers/chemistry , Reactive Oxygen Species/metabolism , Cell Survival/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Astrocytes/metabolism , Astrocytes/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Autophagy/drug effects
2.
PLoS One ; 19(7): e0298102, 2024.
Article in English | MEDLINE | ID: mdl-38954731

ABSTRACT

Brain tumors pose a significant threat to health, and their early detection and classification are crucial. Currently, the diagnosis heavily relies on pathologists conducting time-consuming morphological examinations of brain images, leading to subjective outcomes and potential misdiagnoses. In response to these challenges, this study proposes an improved Vision Transformer-based algorithm for human brain tumor classification. To overcome the limitations of small existing datasets, Homomorphic Filtering, Channels Contrast Limited Adaptive Histogram Equalization, and Unsharp Masking techniques are applied to enrich dataset images, enhancing information and improving model generalization. Addressing the limitation of the Vision Transformer's self-attention structure in capturing input token sequences, a novel relative position encoding method is employed to enhance the overall predictive capabilities of the model. Furthermore, the introduction of residual structures in the Multi-Layer Perceptron tackles convergence degradation during training, leading to faster convergence and enhanced algorithm accuracy. Finally, this study comprehensively analyzes the network model's performance on validation sets in terms of accuracy, precision, and recall. Experimental results demonstrate that the proposed model achieves a classification accuracy of 91.36% on an augmented open-source brain tumor dataset, surpassing the original VIT-B/16 accuracy by 5.54%. This validates the effectiveness of the proposed approach in brain tumor classification, offering potential reference for clinical diagnoses by medical practitioners.


Subject(s)
Algorithms , Brain Neoplasms , Humans , Brain Neoplasms/pathology , Brain Neoplasms/classification , Brain Neoplasms/diagnostic imaging , Neural Networks, Computer
3.
Theranostics ; 14(10): 4107-4126, 2024.
Article in English | MEDLINE | ID: mdl-38994023

ABSTRACT

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


Subject(s)
CCAAT-Enhancer-Binding Protein-beta , Glioblastoma , Tumor Microenvironment , Tumor-Associated Macrophages , Glioblastoma/pathology , Glioblastoma/metabolism , Glioblastoma/genetics , Humans , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , Animals , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Mice , Cell Line, Tumor , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Signal Transduction , Macrophages/metabolism
4.
Front Immunol ; 15: 1384249, 2024.
Article in English | MEDLINE | ID: mdl-38994360

ABSTRACT

Glioblastoma (GBM) tumors are the most aggressive primary brain tumors in adults that, despite maximum treatment, carry a dismal prognosis. GBM tumors exhibit tissue hypoxia, which promotes tumor aggressiveness and maintenance of glioma stem cells and creates an overall immunosuppressive landscape. This article reviews how hypoxic conditions overlap with inflammatory responses, favoring the proliferation of immunosuppressive cells and inhibiting cytotoxic T cell development. Immunotherapies, including vaccines, immune checkpoint inhibitors, and CAR-T cell therapy, represent promising avenues for GBM treatment. However, challenges such as tumor heterogeneity, immunosuppressive TME, and BBB restrictiveness hinder their effectiveness. Strategies to address these challenges, including combination therapies and targeting hypoxia, are actively being explored to improve outcomes for GBM patients. Targeting hypoxia in combination with immunotherapy represents a potential strategy to enhance treatment efficacy.


Subject(s)
Brain Neoplasms , Glioblastoma , Tumor Microenvironment , Humans , Glioblastoma/immunology , Glioblastoma/therapy , Glioblastoma/pathology , Tumor Microenvironment/immunology , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Brain Neoplasms/pathology , Animals , Immunotherapy/methods , Tumor Hypoxia
5.
J Transl Med ; 22(1): 648, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38987805

ABSTRACT

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.


Subject(s)
Cannabidiol , Glioma , Cannabidiol/therapeutic use , Cannabidiol/pharmacology , Humans , Glioma/drug therapy , Glioma/pathology , Animals , Translational Research, Biomedical , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Nanomedicine/methods
6.
Adv Tech Stand Neurosurg ; 52: 73-90, 2024.
Article in English | MEDLINE | ID: mdl-39017787

ABSTRACT

BACKGROUND: Fully endoscopic or endoscope-controlled approaches are essentially keyhole approaches in which rigid endoscopes are the sole visualization tools used during the whole procedure. At the early attempts of endoscope-assisted cranial surgery, it was noted that rigid endoscopes enabled overcoming the problem of suboptimal visualization when small exposures are used. The technical specifications and design of the currently available rigid endoscopes are associated with a group of unique features that define the endoscopic view and lay the basis for its superiority over the microscopic view during brain surgery. Fully endoscopic resection of intraparenchymal brain tumors is a minimally invasive approach that is not routinely practiced by neurosurgeons, with a few major series published so far. Unfamiliarity with the technique, steep learning curve, and concerns about inadequate exposure and decreased visibility may explain this fact. The majority of the purely endoscopic resections for intraparenchymal brain lesions are performed nowadays through tubular retractor systems. In very limited instances, however, the fully endoscopic technique is performed without tubular retractors. In this chapter, we elaborate on the surgical technique and nuances of the fully endoscopic nontubular retractor approach for intraaxial tumors. METHODS: From a prospective database of endoscopic procedures maintained by the senior author, clinical data, imaging studies, and operative charts and videos of cases undergoing fully endoscopic excision for intraaxial brain tumors were retrieved and analyzed. The pertinent literature was also reviewed. RESULTS: The surgical technique of the fully endoscopic nontubular retractor approach for intraaxial tumors was formulated. CONCLUSION: The endoscopic technique has many advantages over the conventional procedures. In our hands, the technique has proven to be feasible, efficient, and minimally invasive with excellent results.


Subject(s)
Brain Neoplasms , Neuroendoscopy , Humans , Brain Neoplasms/surgery , Brain Neoplasms/pathology , Neuroendoscopy/methods , Neuroendoscopy/instrumentation
7.
ACS Appl Mater Interfaces ; 16(28): 35925-35935, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38950334

ABSTRACT

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.


Subject(s)
Glioblastoma , Nanoparticles , Porphyrins , Theranostic Nanomedicine , Glioblastoma/diagnostic imaging , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/metabolism , Animals , Nanoparticles/chemistry , Humans , Porphyrins/chemistry , Porphyrins/pharmacology , Mice , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Infrared Rays , Tissue Distribution , Blood-Brain Barrier/metabolism , Mice, Nude , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Mice, Inbred BALB C , Fluorescence
8.
Cell Rep Med ; 5(7): 101630, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38955178

ABSTRACT

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).


Subject(s)
Brain Neoplasms , Glioma , Humans , Male , Middle Aged , Glioma/drug therapy , Glioma/pathology , Adult , Female , Aged , Administration, Oral , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Aged, 80 and over , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Neoplasm Grading , Maximum Tolerated Dose
9.
Cell Death Dis ; 15(7): 485, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38971772

ABSTRACT

The discovery of novel oncotargets for glioma is of immense significance. We here explored the expression patterns, biological functions, and underlying mechanisms associated with ORC6 (origin recognition complex 6) in glioma. Through the bioinformatics analyses, we found a significant increase in ORC6 expression within human glioma tissues, correlating with poorer overall survival, higher tumor grade, and wild-type isocitrate dehydrogenase status. Additionally, ORC6 overexpression is detected in glioma tissues obtained from locally-treated patients and across various primary/established glioma cells. Further bioinformatics scrutiny revealed that genes co-expressed with ORC6 are enriched in multiple signaling cascades linked to cancer. In primary and immortalized (A172) glioma cells, depleting ORC6 using specific shRNA or Cas9-sgRNA knockout (KO) significantly decreased cell viability and proliferation, disrupted cell cycle progression and mobility, and triggered apoptosis. Conversely, enhancing ORC6 expression via a lentiviral construct augmented malignant behaviors in human glioma cells. ORC6 emerged as a crucial regulator for the expression of key oncogenic genes, including Cyclin A2, Cyclin B2, and DNA topoisomerase II (TOP2A), within glioma cells. Silencing or KO of ORC6 reduced the mRNA and protein levels of these genes, while overexpression of ORC6 increased their expression in primary glioma cells. Bioinformatics analyses further identified RBPJ as a potential transcription factor of ORC6. RBPJ shRNA decreased ORC6 expression in primary glioma cells, while its overexpression increased it. Additionally, significantly enhanced binding between the RBPJ protein and the proposed ORC6 promoter region was detected in glioma tissues and cells. In vivo experiments demonstrated a significant reduction in the growth of patient-derived glioma xenografts in the mouse brain subsequent to ORC6 KO. ORC6 depletion, inhibited proliferation, decreased expression of Cyclin A2/B2/TOP2A, and increased apoptosis were detected within these ORC6 KO intracranial glioma xenografts. Altogether, RBPJ-driven ORC6 overexpression promotes glioma cell growth, underscoring its significance as a promising therapeutic target.


Subject(s)
Cell Proliferation , Gene Expression Regulation, Neoplastic , Glioma , Origin Recognition Complex , Animals , Humans , Male , Mice , Apoptosis/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cyclin A2/metabolism , Cyclin A2/genetics , Cyclin B2/metabolism , Cyclin B2/genetics , DNA Topoisomerases, Type II/metabolism , DNA Topoisomerases, Type II/genetics , Glioma/genetics , Glioma/pathology , Glioma/metabolism , Mice, Nude , Origin Recognition Complex/metabolism , Origin Recognition Complex/genetics
10.
PeerJ ; 12: e17631, 2024.
Article in English | MEDLINE | ID: mdl-39006026

ABSTRACT

Background: Human olfactory receptors (ORs) account for approximately 60% of all human G protein-coupled receptors. The functions of ORs extend beyond olfactory perception and have garnered significant attention in tumor biology. However, a comprehensive pan-cancer analysis of ORs in human cancers is lacking. Methods: Using data from public databases, such as HPA, TCGA, GEO, GTEx, TIMER2, TISDB, UALCAN, GEPIA2, and GSCA, this study investigated the role of olfactory receptor family 7 subfamily A member 5 (OR7A5) in various cancers. Functional analysis of OR7A5 in LGG and GBM was performed using the CGGA database. Molecular and cellular experiments were performed to validate the expression and biological function of OR7A5 in gliomas. Results: The results revealed heightened OR7A5 expression in certain tumors, correlating with the expression levels of immune checkpoints and immune infiltration. In patients with gliomas, the expression levels of OR7A5 were closely associated with adverse prognosis, 1p/19p co-deletion status, and wild-type IDH status. Finally, in vitro experiments confirmed the inhibitory effect of OR7A5 knockdown on the proliferative capacity of glioma cells and on the expression levels of proteins related to lipid metabolism. Conclusion: This study establishes OR7A5 as a novel biomarker, potentially offering a novel therapeutic target for gliomas.


Subject(s)
Biomarkers, Tumor , Brain Neoplasms , Glioma , Receptors, Odorant , Humans , Receptors, Odorant/genetics , Receptors, Odorant/metabolism , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Glioma/immunology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/immunology , Cell Line, Tumor , Prognosis , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic
11.
Front Immunol ; 15: 1420182, 2024.
Article in English | MEDLINE | ID: mdl-39007144

ABSTRACT

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


Subject(s)
Brain Neoplasms , Glioblastoma , Matrix Metalloproteinase 1 , Glioblastoma/pathology , Glioblastoma/metabolism , Glioblastoma/genetics , Humans , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 1/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Cell Line, Tumor , Cell Proliferation , Cell Movement/genetics , Disease Progression , Inflammation/metabolism , Gene Expression Regulation, Neoplastic , Signal Transduction , Male
12.
Neurology ; 103(3): e209688, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39008801

ABSTRACT

The discovery in 2008 that many adult gliomas harbor a hitherto unknown mutation in the metabolic gene isocitrate dehydrogenase (IDH) initiated revolutionary advances in our understanding of the biology, and correspondingly our classification, of gliomas. IDH mutations are found in most nonglioblastoma adult gliomas and portend a better prognosis. Massive efforts have unraveled many of the pleiotropic cellular effects of these mutations and spawned several lines of investigation to target the effect to therapeutic benefit. In this article are reviewed the implications of the IDH mutation in gliomas, in particular focusing on recent studies that have culminated in a rare positive phase 3 trial in these generally refractory tumors.


Subject(s)
Brain Neoplasms , Glioma , Isocitrate Dehydrogenase , Mutation , Humans , Glioma/genetics , Glioma/therapy , Isocitrate Dehydrogenase/genetics , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Brain Neoplasms/pathology , Molecular Targeted Therapy
13.
J Vis Exp ; (208)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39007604

ABSTRACT

Pineal neoplasms have a significant impact on children although they are relatively uncommon. They account for approximately 3-11% of all childhood brain tumors, which is considerably higher than the <1% seen in adult brain tumors. These tumors can be divided into three main categories: germ cell tumors, parenchymal pineal tumors, and tumors arising from related anatomical structures. Obtaining an accurate and minimally invasive tissue diagnosis is crucial for selecting the most appropriate treatment regimen for patients with pineal gland tumors. This is due to the diverse treatment options available and the potential risks associated with complete resection. In cases where patients present with acute obstructive hydrocephalus caused by a pineal gland tumor, immediate treatment of the hydrocephalus is necessary. The urgency stems from the potential complications of hydrocephalus, including increased intracranial pressure and neurological deficits. To address these challenges, a minimally invasive endoscopic approach provides a valuable opportunity. This technique allows clinicians to promptly relieve hydrocephalus and obtain a histological diagnosis simultaneously. This dual benefit enables a more comprehensive understanding of the tumor and assists in determining the most effective treatment strategy for the patient.


Subject(s)
Brain Neoplasms , Pineal Gland , Pinealoma , Ventriculostomy , Humans , Ventriculostomy/methods , Pineal Gland/surgery , Pineal Gland/pathology , Pinealoma/surgery , Pinealoma/pathology , Brain Neoplasms/surgery , Brain Neoplasms/pathology , Biopsy/methods , Hydrocephalus/surgery , Hydrocephalus/pathology , Third Ventricle/surgery , Third Ventricle/pathology , Neuroendoscopy/methods
14.
Cells ; 13(13)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38995006

ABSTRACT

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.


Subject(s)
Aurora Kinase A , B7-H1 Antigen , Glioblastoma , Killer Cells, Natural , Aurora Kinase A/metabolism , Aurora Kinase A/antagonists & inhibitors , Humans , Glioblastoma/pathology , Glioblastoma/drug therapy , Glioblastoma/immunology , Glioblastoma/genetics , B7-H1 Antigen/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Animals , Mice , Cell Line, Tumor , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Azepines/pharmacology , Pyrimidines/pharmacology , Cytotoxicity, Immunologic/drug effects , Brain Neoplasms/pathology , Brain Neoplasms/immunology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Xenograft Model Antitumor Assays
15.
Cells ; 13(13)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38994929

ABSTRACT

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


Subject(s)
Glioblastoma , Receptors, Chimeric Antigen , Tumor Microenvironment , Glioblastoma/therapy , Glioblastoma/immunology , Glioblastoma/radiotherapy , Glioblastoma/pathology , Animals , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Mice , Tumor Microenvironment/immunology , Humans , Cell Line, Tumor , Immunotherapy, Adoptive/methods , Brain Neoplasms/therapy , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , T-Lymphocytes/immunology , Mice, Inbred C57BL , Immunomodulation , Female
16.
Medicine (Baltimore) ; 103(28): e38913, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38996177

ABSTRACT

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


Subject(s)
Biomarkers, Tumor , Brain Neoplasms , Glioblastoma , Glycoproteins , Intercellular Signaling Peptides and Proteins , Humans , Glioblastoma/metabolism , Glioblastoma/mortality , Glioblastoma/genetics , Glioblastoma/pathology , Female , Male , Middle Aged , Biomarkers, Tumor/metabolism , Glycoproteins/metabolism , Prognosis , Aged , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Adult , Temozolomide/therapeutic use , Kaplan-Meier Estimate , Progression-Free Survival , Retrospective Studies , Antineoplastic Agents, Alkylating/therapeutic use , Immunohistochemistry
17.
CNS Neurosci Ther ; 30(7): e14791, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38997808

ABSTRACT

INTRODUCTION: Glioblastoma (GBM) remains a challenging brain tumor to treat, with limited response to PD-1 immunotherapy due to tumor-associated macrophages (TAMs), specifically the M2 phenotype. This study explores the potential of MS4A4A (membrane spanning four domains, subfamily A, member 4A) inhibition in driving M2 macrophage polarization toward the M1 phenotype via the ferroptosis pathway to enhance the effectiveness of immunotherapy in GBM. METHODS: Single-cell RNA sequencing and spatial transcriptomic analyses were employed to characterize M2 macrophages and MS4A4A expression in GBM. In vitro studies utilizing TAM cultures, flow cytometry, and western blot validations were conducted to assess the impact of MS4A4A on the tumor immune microenvironment and M2 macrophage polarization. In vivo models, including subcutaneous and orthotopic transplantation in mice, were utilized to evaluate the effects of MS4A4A knockout and combined immune checkpoint blockade (ICB) therapy on tumor growth and response to PD-1 immunotherapy. RESULTS: Distinct subsets of GBM-associated macrophages were identified, with spatial distribution in tumor tissue elucidated. In vivo experiments demonstrated that inhibiting MS4A4A and combining ICB therapy effectively inhibited tumor growth, reshaped the tumor immune microenvironment by reducing M2 TAM infiltration and enhancing CD8+ T-cell infiltration, ultimately leading to complete tumor eradication. CONCLUSION: MS4A4A inhibition shows promise in converting M2 macrophages to M1 phenotype via ferroptosis, decreasing M2-TAM infiltration, and enhancing GBM response to PD-1 immunotherapy. These findings offer a novel approach to developing more effective immunotherapeutic strategies for GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , Immunotherapy , Glioblastoma/immunology , Glioblastoma/therapy , Glioblastoma/pathology , Animals , Immunotherapy/methods , Mice , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Brain Neoplasms/pathology , Humans , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor Microenvironment/physiology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/drug effects , Mice, Inbred C57BL , Cell Line, Tumor , Membrane Proteins/metabolism , Membrane Proteins/genetics
18.
Sci Rep ; 14(1): 16031, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992201

ABSTRACT

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


Subject(s)
Brain Neoplasms , DNA Methylation , DNA Modification Methylases , DNA Repair Enzymes , Glioblastoma , Magnetic Resonance Imaging , Promoter Regions, Genetic , Tumor Suppressor Proteins , Humans , Glioblastoma/genetics , Glioblastoma/diagnostic imaging , Glioblastoma/pathology , DNA Repair Enzymes/genetics , DNA Modification Methylases/genetics , Tumor Suppressor Proteins/genetics , Magnetic Resonance Imaging/methods , Female , Male , Middle Aged , Brain Neoplasms/genetics , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Retrospective Studies , Adult , Aged , Prognosis , ROC Curve , Radiomics
19.
Bull Exp Biol Med ; 177(1): 147-154, 2024 May.
Article in English | MEDLINE | ID: mdl-38963598

ABSTRACT

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.


Subject(s)
Brain Neoplasms , Doxorubicin , Glioblastoma , Hyaluronoglucosaminidase , Polyethylene Glycols , Animals , Doxorubicin/pharmacology , Hyaluronoglucosaminidase/metabolism , Rats , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Glioblastoma/drug therapy , Glioblastoma/pathology , Male , Cell Line, Tumor , Spheroids, Cellular/drug effects
20.
Nat Biomed Eng ; 8(6): 672-688, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38987630

ABSTRACT

The most widely used fluorophore in glioma-resection surgery, 5-aminolevulinic acid (5-ALA), is thought to cause the selective accumulation of fluorescent protoporphyrin IX (PpIX) in tumour cells. Here we show that the clinical detection of PpIX can be improved via a microscope that performs paired stimulated Raman histology and two-photon excitation fluorescence microscopy (TPEF). We validated the technique in fresh tumour specimens from 115 patients with high-grade gliomas across four medical institutions. We found a weak negative correlation between tissue cellularity and the fluorescence intensity of PpIX across all imaged specimens. Semi-supervised clustering of the TPEF images revealed five distinct patterns of PpIX fluorescence, and spatial transcriptomic analyses of the imaged tissue showed that myeloid cells predominate in areas where PpIX accumulates in the intracellular space. Further analysis of external spatially resolved metabolomics, transcriptomics and RNA-sequencing datasets from glioblastoma specimens confirmed that myeloid cells preferentially accumulate and metabolize PpIX. Our findings question 5-ALA-induced fluorescence in glioma cells and show how 5-ALA and TPEF imaging can provide a window into the immune microenvironment of gliomas.


Subject(s)
Brain Neoplasms , Glioma , Protoporphyrins , Spectrum Analysis, Raman , Protoporphyrins/metabolism , Humans , Glioma/pathology , Glioma/metabolism , Glioma/surgery , Glioma/diagnostic imaging , Spectrum Analysis, Raman/methods , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/surgery , Brain Neoplasms/diagnostic imaging , Microscopy, Fluorescence/methods , Aminolevulinic Acid/metabolism , Female , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...