Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 330
Filter
1.
Biomed Khim ; 70(2): 114-124, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38711411

ABSTRACT

Existing knowledge on changes of the haptoglobin (Hp) molecule suggests that it may exist in multiple proteoforms, which obviously exhibit different functions. Using two-dimensional electrophoresis (2DE) in combination with mass spectrometry and immunodetection, we have analyzed blood plasma samples from both healthy donors and patients with primary grade IV glioblastoma (GBM), and obtained a detailed composite 2DE distribution map of ß-chain proteoforms, as well as the full-length form of Hp (zonulin). Although the total level of plasma Hp exceeded normal values in cancer patients (especially patients with GBM), the presence of particuar proteoforms, detected by their position on the 2DE map, was very individual. Variability was found in both zonulin and the Hp ß-chain. The presence of an alkaline form of zonulin in plasma can be considered a conditional, but insufficient, GBM biomarker. In other words, we found that at the level of minor proteoforms of Hp, even in normal conditions, there was a high individual variability. On the one hand, this raises questions about the reasons for such variability, if it is present not only in Hp, but also in other proteins. On the other hand, this may explain the discrepancy between the number of experimentally detected proteoforms and the theoretically possible ones not only in Hp, but also in other proteins.


Subject(s)
Glioblastoma , Haptoglobins , Protein Precursors , Haptoglobins/analysis , Haptoglobins/metabolism , Haptoglobins/chemistry , Humans , Female , Male , Glioblastoma/blood , Glioblastoma/metabolism , Middle Aged , Biomarkers, Tumor/blood , Aged , Electrophoresis, Gel, Two-Dimensional/methods , Adult
2.
Aging (Albany NY) ; 16(9): 8306-8319, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38742944

ABSTRACT

BACKGROUND: Glioblastoma Multiforme (GBM) is one of the most aggressive and fatal brain cancers. The study of metabolites could be crucial for understanding GBM's biology and reveal new treatment strategies. METHODS: The GWAS data for GBM were sourced from the FinnGen database. A total of 1400 plasma metabolites were collected from the GWAS Catalog dataset. The cerebrospinal fluid (CSF) metabolites data were collected from subsets of participants in the WADRC and WRAP studies. We utilized the inverse variance weighting (IVW) method as the primary tool to explore the causal relationship between metabolites in plasma and CSF and glioblastoma, ensuring the exclusion of instances with horizontal pleiotropy. Additionally, four supplementary analytical methods were applied to reinforce our findings. Aberrant results were identified and omitted based on the outcomes of the leave-one-out sensitivity analysis. Conclusively, a reverse Mendelian Randomization analysis was also conducted to further substantiate our results. RESULTS: The study identified 69 plasma metabolites associated with GBM. Of these, 40 metabolites demonstrated a significant positive causal relationship with GBM, while 29 exhibited a significant negative causal association. Notably, Trimethylamine N-oxide (TMAO) levels in plasma, not CSF, were found to be a significant exposure factor for GBM (OR = 3.1627, 95% CI = (1.6347, 6.1189), P = 0.0006). The study did not find a reverse causal relationship between GBM and plasma TMAO levels. CONCLUSIONS: This research has identified 69 plasma metabolites potentially associated with the incidence of GBM, among which TMAO stands out as a promising candidate for an early detectable biomarker for GBM.


Subject(s)
Brain Neoplasms , Genome-Wide Association Study , Glioblastoma , Mendelian Randomization Analysis , Humans , Glioblastoma/cerebrospinal fluid , Glioblastoma/blood , Glioblastoma/genetics , Brain Neoplasms/cerebrospinal fluid , Brain Neoplasms/genetics , Brain Neoplasms/blood , Biomarkers, Tumor/cerebrospinal fluid , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Methylamines/blood , Methylamines/cerebrospinal fluid , Female , Male
3.
Int J Mol Sci ; 25(8)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38673808

ABSTRACT

Novel blood-circulating molecules, as potential biomarkers for glioblastoma multiforme (GBM) diagnosis and monitoring, are attracting particular attention due to limitations of imaging modalities and invasive tissue biopsy procedures. This study aims to assess the diagnostic and prognostic values of circulating cell-free DNA (cfDNA) in relation to inflammatory status in GBM patients and to determine the concentration and average size of DNA fragments typical of tumour-derived DNA fractions. Preoperative plasma samples from 40 patients (GBM 65.0 ± 11.3 years) and 40 healthy controls (HC 70.4 ± 5.4 years) were compared. The cfDNA concentrations and lengths were measured using the electrophoresis platform, and inflammatory indices (NLR, PLR, LMR, and SII) were calculated from complete blood cell analysis. More fragmented cfDNA and 4-fold higher 50-700 bp cfDNA concentrations were detected in GBM patients than in healthy controls. The average cfDNA size in the GBM group was significantly longer (median 336 bp) than in the HC group (median 271 bp). Optimal threshold values were 1265 pg/µL for 50-700 bp cfDNA (AUC = 0.857) and 290 bp for average cfDNA size (AUC = 0.814). A Kaplan-Meier survival curves analysis also demonstrated a higher mortality risk in the GBM group with a cut-off >303 bp cfDNA. This study is the first to have revealed glioblastoma association with high levels of cfDNA > 1000 pg/µL of 50-700 bp in length, which can be aggravated by immunoinflammatory reactivity.


Subject(s)
Biomarkers, Tumor , Cell-Free Nucleic Acids , Glioblastoma , Humans , Glioblastoma/blood , Glioblastoma/diagnosis , Glioblastoma/mortality , Glioblastoma/genetics , Male , Female , Aged , Middle Aged , Prognosis , Biomarkers, Tumor/blood , Cell-Free Nucleic Acids/blood , Brain Neoplasms/blood , Brain Neoplasms/diagnosis , Brain Neoplasms/mortality , Kaplan-Meier Estimate , Case-Control Studies , Circulating Tumor DNA/blood
4.
Sci Rep ; 11(1): 22792, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34815502

ABSTRACT

Glioblastoma is a devastating tumor of the central nervous system characterized by a poor survival and an extremely dark prognosis, making its diagnosis, treatment and monitoring highly challenging. Numerous studies have highlighted extracellular vesicles (EVs) as key players of tumor growth, invasiveness and resistance, as they carry and disseminate oncogenic material in the local tumor microenvironment and at distance. However, whether their quality and quantity reflect individual health status and changes in homeostasis is still not fully elucidated. Here, we separated EVs from plasma collected at different time points alongside with the clinical management of GBM patients. Our findings confirm that plasmatic EVs could be separated and characterized with standardized protocols, thereby ensuring the reliability of measuring vesiclemia, i.e. extracellular vesicle concentration in plasma. This unveils that vesiclemia is a dynamic parameter, which could be reflecting tumor burden and/or response to treatments. Further label-free liquid chromatography tandem mass spectrometry unmasks the von Willebrand Factor (VWF) as a selective protein hallmark for GBM-patient EVs. Our data thus support the notion that EVs from GBM patients showed differential protein cargos that can be further surveyed in circulating EVs, together with vesiclemia.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , Extracellular Vesicles/metabolism , Glioblastoma/pathology , Proteome/metabolism , Tumor Microenvironment/immunology , von Willebrand Factor/metabolism , Aged , Aged, 80 and over , Brain Neoplasms/blood , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Case-Control Studies , Female , Follow-Up Studies , Glioblastoma/blood , Glioblastoma/immunology , Glioblastoma/metabolism , Humans , Male , Middle Aged , Prognosis , Proteome/analysis
5.
J Neurooncol ; 155(2): 173-180, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34652553

ABSTRACT

PURPOSE: Up to 30% of patients with glioblastoma (GBM) develop venous thromboembolism (VTE) over the course of the disease. Although not as high, the risk for VTE is also increased in patients with meningioma. Direct measurement of peak thrombin generation (TG) allows quantitative assessment of systemic coagulation activation in patients with GBM and meningioma. Our aim was to determine the extent of systemic coagulation activation induced by brain tumors, to measure the shift between pre- and post-operative peak TG in patients with GBM, and to assess the relationship between pre-surgical peak TG and pre-operative brain tumor volume on imaging. METHODS: Pre- and post-surgical plasma samples were obtained from successive patients with GBM and once from patients with meningioma and healthy age- and sex-matched blood donor controls. TG was measured using the calibrated automated thrombogram (CAT) assay, and tumor volumes were measured in pre-surgical MRI scans. RESULTS: Pre-surgical peak TG was higher in patients with GBM than in controls (288.6 ± 54.1 nM vs 187.1 ± 41.7 nM, respectively, P < 0.001), and, in the nine patients with GBM and paired data available, peak TG was significantly reduced after surgery (323 ± 38 nM vs 265 ± 52 nM, respectively, P = 0.007). Similarly, subjects with meningioma demonstrated higher peak TG compared to controls (242.2 ± 54.9 nM vs 177.7 ± 57.0 nM, respectively, P < 0.001). There was no association between peak TG and pre-operative tumor volume or overall survival. CONCLUSION: Our results indicate that systemic coagulation activation occurs with both meningioma and GBM, but to a greater degree in the latter. Preoperative peak TG did not correlate with tumor volume, but removal of GBM caused a significant decrease in coagulation activation.


Subject(s)
Blood Coagulation , Brain Neoplasms , Glioblastoma , Meningeal Neoplasms , Meningioma , Blood Coagulation/physiology , Brain Neoplasms/blood , Glioblastoma/blood , Humans , Meningeal Neoplasms/blood , Meningioma/blood
6.
Molecules ; 26(19)2021 Oct 02.
Article in English | MEDLINE | ID: mdl-34641541

ABSTRACT

Glioblastoma Multiforme (GBM) is a brain tumor with a poor prognosis and low survival rates. GBM is diagnosed at an advanced stage, so little information is available on the early stage of the disease and few improvements have been made for earlier diagnosis. Longitudinal murine models are a promising platform for biomarker discovery as they allow access to the early stages of the disease. Nevertheless, their use in proteomics has been limited owing to the low sample amount that can be collected at each longitudinal time point. Here we used optimized microproteomics workflows to investigate longitudinal changes in the protein profile of serum, serum small extracellular vesicles (sEVs), and cerebrospinal fluid (CSF) in a GBM murine model. Baseline, pre-symptomatic, and symptomatic tumor stages were determined using non-invasive motor tests. Forty-four proteins displayed significant differences in signal intensities during GBM progression. Dysregulated proteins are involved in cell motility, cell growth, and angiogenesis. Most of the dysregulated proteins already exhibited a difference from baseline at the pre-symptomatic stage of the disease, suggesting that early effects of GBM might be detectable before symptom onset.


Subject(s)
Brain Neoplasms/blood , Brain Neoplasms/cerebrospinal fluid , Glioblastoma/blood , Glioblastoma/cerebrospinal fluid , Proteomics/methods , Animals , Biomarkers, Tumor/blood , Biomarkers, Tumor/cerebrospinal fluid , Blood Proteins/analysis , Cerebrospinal Fluid Proteins/analysis , Extracellular Vesicles/pathology , Female , Male , Mice, Inbred C57BL , Neoplasms, Experimental/blood , Neoplasms, Experimental/cerebrospinal fluid , Neoplasms, Experimental/pathology , Workflow
7.
Theranostics ; 11(19): 9217-9233, 2021.
Article in English | MEDLINE | ID: mdl-34646367

ABSTRACT

Tryptophan (Trp)-catabolic enzymes (TCEs) produce metabolites that activate the aryl hydrocarbon receptor (AHR) and promote tumor progression and immunosuppression in glioblastoma. As therapies targeting TCEs or AHR become available, a better understanding of Trp metabolism is required. Methods: The combination of LC-MS/MS with chemical isobaric labeling enabled the simultaneous quantitative comparison of Trp and its amino group-bearing metabolites in multiple samples. We applied this method to the sera of a cohort of 43 recurrent glioblastoma patients and 43 age- and sex-matched healthy controls. Tumor volumes were measured in MRI data using an artificial neural network-based approach. MALDI MSI visualized Trp and its direct metabolite N-formylkynurenine (FK) in glioblastoma tissue. Analysis of scRNA-seq data was used to detect the presence of Trp metabolism and AHR activity in different cell types in glioblastoma. Results: Compared to healthy controls, glioblastoma patients showed decreased serum Trp levels. Surprisingly, the levels of Trp metabolites were also reduced. The decrease became smaller with more enzymatic steps between Trp and its metabolites, suggesting that Trp availability controls the levels of its systemic metabolites. High tumor volume associated with low systemic metabolite levels and low systemic kynurenine levels associated with worse overall survival. MALDI MSI demonstrated heterogeneity of Trp catabolism across glioblastoma tissues. Analysis of scRNA-seq data revealed that genes involved in Trp metabolism were expressed in almost all the cell types in glioblastoma and that most cell types, in particular macrophages and T cells, exhibited AHR activation. Moreover, high AHR activity associated with reduced overall survival in the glioblastoma TCGA dataset. Conclusion: The novel techniques we developed could support the identification of patients that may benefit from therapies targeting TCEs or AHR activation.


Subject(s)
Glioblastoma/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Tryptophan/metabolism , Cell Line, Tumor , Chromatography, Liquid/methods , Cohort Studies , Databases, Genetic , Female , Glioblastoma/blood , Glioblastoma/genetics , Humans , Immunotherapy , Male , Middle Aged , Receptors, Aryl Hydrocarbon/genetics , Tandem Mass Spectrometry/methods , Tryptophan/blood
8.
J Neuroimmunol ; 361: 577743, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34695769

ABSTRACT

We present a patient with positive medium titer MOG-IgG and progressive neurological decline whose clinical and radiological phenotype were not consistent with a MOG-IgG associated disorder and ultimately received a diagnosis of glioblastoma after brain biopsy and died 4 weeks later. This represents an important topic with a high frequency of MOG-IgG testing in clinical practice. Due to this there are increasing reports of MOG-IgG positivity in atypical clinical phenotypes, raising the possibility of false positives, which can have important implications. It is important to highlight that judicious clinical evaluation is needed when interpreting MOG-IgG results in atypical settings.


Subject(s)
Antibodies, Neoplasm/blood , Autoantibodies/blood , Autoantigens/immunology , Brain Neoplasms/immunology , Diagnostic Errors , Glioblastoma/immunology , Immunoglobulin G/blood , Myelin-Oligodendrocyte Glycoprotein/immunology , Adult , Autoantibodies/immunology , Biopsy , Brain Neoplasms/blood , Brain Neoplasms/diagnostic imaging , Delayed Diagnosis , Facial Paralysis/etiology , False Positive Reactions , Female , Gait Disorders, Neurologic/etiology , Glioblastoma/blood , Glioblastoma/diagnostic imaging , Humans , Immunoglobulin G/immunology , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Neuroimaging , Paresis/etiology , Positron-Emission Tomography , Vertigo/etiology , Vision Disorders/etiology
9.
PLoS One ; 16(9): e0256831, 2021.
Article in English | MEDLINE | ID: mdl-34495981

ABSTRACT

Current approach for the detection of cancer is based on identifying genetic mutations typical to tumor cells. This approach is effective only when cancer has already emerged, however, it might be in a stage too advanced for effective treatment. Cancer is caused by the continuous accumulation of mutations; is it possible to measure the time-dependent information of mutation accumulation and predict the emergence of cancer? We hypothesize that the mutation history derived from the tandem repeat regions in blood-derived DNA carries information about the accumulation of the cancer driver mutations in other tissues. To validate our hypothesis, we computed the mutation histories from the tandem repeat regions in blood-derived exomic DNA of 3874 TCGA patients with different cancer types and found a statistically significant signal with specificity ranging from 66% to 93% differentiating Glioblastoma patients from other cancer patients. Our approach and findings offer a new direction for future cancer prediction and early cancer detection based on information derived from blood-derived DNA.


Subject(s)
Blood Cells/metabolism , Brain Neoplasms/blood , Brain Neoplasms/genetics , DNA/genetics , Early Detection of Cancer/methods , Glioblastoma/blood , Glioblastoma/genetics , Mutation , Brain Neoplasms/pathology , Databases, Genetic , Exome , Glioblastoma/pathology , Humans , Machine Learning , Sensitivity and Specificity , Sequence Alignment/methods , Tandem Repeat Sequences/genetics
10.
Cancer Med ; 10(15): 5154-5162, 2021 08.
Article in English | MEDLINE | ID: mdl-34250760

ABSTRACT

BACKGROUND: Investigation of novel blood-circulating agents as potential biomarkers for glioblastoma multiforme (GBM) patients' diagnosis and monitoring has gained lots of attention, due to limitations of imaging modalities and invasive tissue biopsy procedures. The present study aims to assess the diagnostic and prognostic values of preoperative stem cell factor (SCF) plasma level in GBM patients. METHODS: Preoperative plasma samples from 58 GBM patients and 20 patients with nonglial tumors and 30 healthy controls were obtained. SCF levels were measured by employing the enzyme-linked immunosorbent assay test and the values were compared between these three groups. Then, the association of SCF plasma level and tumor volume, progression-free survival (PFS), and overall survival (OS) for the GBM patients were evaluated. RESULTS: Mean preoperative SCF plasma level of the GBM patients (2.80 ± 1.52 ng/ml) was significantly higher (p < 0.0001) than the healthy controls (0.80 ± 0.24 ng/ml) and patients with nonglial tumor (1.41 ± 0.76 ng/ml). Receiver operating characteristic analysis revealed that the preoperative SCF plasma level could distinguish the GBM patients from healthy controls and patients with nonglial tumors with the area under curve values of 0.915 and 0.790, respectively. However, no significant association was observed between the GBM patients' preoperative SCF plasma levels and tumors' volume (Spearman Rho correlation coefficient, 0.1847; 95% CI, p = 0.1652). The GBM patients were divided into two subgroups based on mean preoperative SCF plasma levels (2.80 ng/ml). No significant difference was observed between the patients' PFS (p = 0.3792) and OS (p = 0.1469) at these two subgroups. CONCLUSION: Taking together, the SCF plasma level can serve as a novel diagnostic blood-circulating biomarker for patients with GBM. However, its plasma level is not correlated with GBM patients' tumor volume, PFS, or OS.


Subject(s)
Biomarkers, Tumor/blood , Brain Neoplasms/blood , Glioblastoma/blood , Stem Cell Factor/blood , Adult , Aged , Aged, 80 and over , Brain Neoplasms/diagnosis , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Case-Control Studies , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Diagnosis, Differential , Enzyme-Linked Immunosorbent Assay , Female , Glioblastoma/diagnosis , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Isocitrate Dehydrogenase/genetics , Male , Methylation , Middle Aged , Mutation , Preoperative Period , Prognosis , Progression-Free Survival , ROC Curve , Tumor Burden , Tumor Suppressor Proteins/metabolism
11.
Immunohorizons ; 5(6): 395-409, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34103370

ABSTRACT

Clinical use of various forms of immunotherapeutic drugs in glioblastoma (GBM), has highlighted severe T cell dysfunction such as exhaustion in GBM patients. However, reversing T cell exhaustion using immune checkpoint inhibitors in GBM clinical trials has not shown significant overall survival benefit. Phenotypically, CD8+ T cells with downregulated CD28 coreceptors, low CD27 expression, increased CD57 expression, and telomere shortening are classified as senescent T cells. These senescent T cells are normally seen as part of aging and also in many forms of solid cancers. Absence of CD28 on T cells leads to several functional irregularities including reduced TCR diversity, incomplete activation of T cells, and defects in Ag-induced proliferation. In the context of GBM, presence and/or function of these CD8+CD28- T cells is unknown. In this clinical correlative study, we investigated the effect of aging as well as tumor microenvironment on CD8+ T cell phenotype as an indicator of its function in GBM patients. We systematically analyzed and describe a large population of CD8+CD28- T cells in both the blood and tumor-infiltrating lymphocytes of GBM patients. We found that phenotypically these CD8+CD28- T cells represent a distinct population compared with exhausted T cells. Comparative transcriptomic and pathway analysis of CD8+CD28- T cell populations in GBM patients revealed that tumor microenvironment might be influencing several immune related pathways and thus further exaggerating the age associated immune dysfunction in this patient population.


Subject(s)
Aging/immunology , Brain Neoplasms/drug therapy , CD8-Positive T-Lymphocytes/immunology , Glioblastoma/drug therapy , Immune Checkpoint Inhibitors/pharmacology , Adult , Aged , Aged, 80 and over , Aging/blood , Brain Neoplasms/blood , Brain Neoplasms/immunology , Brain Neoplasms/pathology , CD28 Antigens/analysis , CD28 Antigens/immunology , CD28 Antigens/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cellular Senescence/immunology , Drug Resistance, Neoplasm , Female , Glioblastoma/blood , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immunophenotyping , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Middle Aged , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Young Adult
12.
Int J Mol Sci ; 22(10)2021 May 11.
Article in English | MEDLINE | ID: mdl-34064637

ABSTRACT

(1) Background: Glioblastoma multiforme (GBM) is among the most aggressive cancers with a poor prognosis. Treatment options are limited, clinicians lack efficient prognostic and predictive markers. Circulating miRNAs-besides being important regulators of cancer development-may have potential as diagnostic biomarkers of GBM. (2) Methods: In this study, profiling of 798 human miRNAs was performed on blood plasma samples from 6 healthy individuals and 6 patients with GBM, using a NanoString nCounter Analysis System. To validate our results, five miRNAs (hsa-miR-433-3p, hsa-miR-362-3p, hsa-miR-195-5p, hsa-miR-133a-3p, and hsa-miR-29a-3p) were randomly chosen for RT-qPCR detection. (3) Results: In all, 53 miRNAs were significantly differentially expressed in plasma samples of GBM patients when data were filtered for FC 1 and FDR 0.1. Target genes of the top 39 differentially expressed miRNAs were identified, and we carried out functional annotation and pathway enrichment analysis of target genes via GO and KEGG-based tools. General and cortex-specific protein-protein interaction networks were constructed from the target genes of top miRNAs to assess their functional connections. (4) Conclusions: We demonstrated that plasma microRNA profiles are promising diagnostic and prognostic molecular biomarkers that may find an actual application in the clinical practice of GBM, although more studies are needed to validate our results.


Subject(s)
Biomarkers, Tumor/genetics , Circulating MicroRNA/genetics , Gene Regulatory Networks , Glioblastoma/genetics , Glioblastoma/pathology , Biomarkers, Tumor/metabolism , Case-Control Studies , Circulating MicroRNA/metabolism , Computational Biology , Gene Expression Profiling , Glioblastoma/blood , Humans , Prognosis , Protein Interaction Maps
13.
Cancer Immunol Immunother ; 70(12): 3643-3650, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33956203

ABSTRACT

PURPOSE: In primary brain tumors, the efficacy of immune-modulating therapies is still under investigation as inflammatory responses are restricted by tight immunoregulatory mechanisms in the central nervous system. Here, we measured soluble PD-L1 (sPD-L1) in the plasma of patients with recurrent glioblastoma (GBM) and recurrent WHO grade II-III glioma treated with bevacizumab-based salvage therapy. METHODS: Thirty patients with recurrent GBM and 10 patients with recurrent WHO grade II-III glioma were treated with bevacizumab-based salvage therapy at the Medical University of Vienna. Prior to each treatment cycle, EDTA plasma was drawn and sPD-L1 was measured applying a sandwich ELISA with a lower detection limit of 0.050 ng/ml. Leukocyte counts and C-reactive protein (CRP) levels were measured according to institutional practice. RESULTS: Median number of sPD-L1 measurements was 6 per patient (range: 2-24). At baseline, no significant difference in sPD-L1 concentrations was observed between WHO grade II-III glioma and GBM. Intra-patient variability of sPD-L1 concentrations was significantly higher in WHO grade II-III glioma than in GBM (p = 0.014) and tendentially higher in IDH-mutant than in IDH-wildtype glioma (p = 0.149) In WHO grade II-III glioma, sPD-L1 levels were significantly lower after one administration of bevacizumab than at baseline (median: 0.039 ng/ml vs. 0.4855 ng/ml, p = 0.036). In contrast, no significant change could be observed in patients with GBM. CONCLUSIONS: Changes in systemic inflammation markers including sPD-L1 are observable in patients with recurrent glioma under bevacizumab-based treatment and differ between WHO grade II-III glioma and GBM.


Subject(s)
B7-H1 Antigen/blood , Bevacizumab/therapeutic use , Glioma/blood , Glioma/drug therapy , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/drug therapy , Adult , Aged , Biomarkers, Tumor/blood , Brain Neoplasms/blood , Brain Neoplasms/drug therapy , Female , Glioblastoma/blood , Glioblastoma/drug therapy , Humans , Male , Middle Aged , Young Adult
14.
Int J Mol Sci ; 22(7)2021 Apr 06.
Article in English | MEDLINE | ID: mdl-33917598

ABSTRACT

Magnetic resonance imaging (MRI) is the gold standard for glioblastoma (GBM) patient evaluation. Additional non-invasive diagnostic modalities are needed. GBM is heavily infiltrated with tumor-associated macrophages (TAMs) that can be found in peripheral blood. FKBP51s supports alternative-macrophage polarization. Herein, we assessed FKBP51s expression in circulating monocytes from 14 GBM patients. The M2 monocyte phenotype was investigated by qPCR and flow cytometry using antibodies against PD-L1, CD163, FKBP51s, and CD14. MRI assessed morphologic features of the tumors that were aligned to flow cytometry data. PD-L1 expression on circulating monocytes correlated with MRI tumor necrosis score. A wider expansion in circulating CD163/monocytes was measured. These monocytes resulted in a dramatic decrease in patients with an MRI diagnosis of complete but not partial surgical removal of the tumor. Importantly, in patients with residual tumor, most of the peripheral monocytes that in the preoperative stage were CD163/FKBP51s- had turned into CD163/FKBP51s+. After Stupp therapy, CD163/FKBP51s+ monocytes were almost absent in a case of pseudoprogression, while two patients with stable or true disease progression showed sustained levels in such circulating monocytes. Our work provides preliminary but meaningful and novel results that deserve to be confirmed in a larger patient cohort, in support of potential usefulness in GBM monitoring of CD163/FKBP51s/CD14 immunophenotype in adjunct to MRI.


Subject(s)
Brain Neoplasms , Flow Cytometry , Glioblastoma , Magnetic Resonance Imaging , Monocytes/metabolism , Real-Time Polymerase Chain Reaction , Adult , Aged , Antigens, CD/blood , Antigens, Differentiation, Myelomonocytic/blood , B7-H1 Antigen/blood , Brain Neoplasms/blood , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/therapy , Female , Glioblastoma/blood , Glioblastoma/diagnostic imaging , Glioblastoma/therapy , Humans , Lipopolysaccharide Receptors/blood , Male , Middle Aged , Prospective Studies , Receptors, Cell Surface/blood , Tacrolimus Binding Proteins/blood
15.
J Clin Neurosci ; 86: 260-266, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33775339

ABSTRACT

PURPOSE: The Controlling Nutritional Status (CONUT) Score, a new parameter that reflects the immuno-nutritional status, has been closely associated with prognosis in many cancer types. However, the prognostic significance of the CONUT score in Glioblastoma Multiforme (GBM) is not known. In this study, we aimed to show the prognostic significance of the CONUT score in the postoperative period in patients with GBM. METHODS: 120 patients followed up with GBM were included in the study, retrospectively. According to the receiver operating characteristic (ROC) curve analysis, the optimal cut-off values were determined for the CONUT score, and the patients were divided into low (<2.5) and high (≥2.5) CONUT groups. Systemic immune inflammation index (SII), prognostic nutritional index (PNI), and neutrophil-lymphocyte ratio (NLR) were grouped according to the cut-off point of 1111, 46.5, and 4.48, respectively. Cox regression analyzes were used to assess their prognostic significance for progression-free survival (PFS) and overall survival (OS). RESULTS: The high CONUT score group was found to have worse PFS and OS than the low CONUT score group (p < 0.001, p < 0.001). In univariate analysis, age, gender, comorbidity, CONUT score, SII, PNI, NLR were found to be significant for both PFS and OS. In multivariate analysis, only age and CONUT score were found as independent prognostic factors for both PFS (p: 0.040, p < 0,001) and OS (p: 0,041, p < 0,001). CONCLUSION: The CONUT score in the postoperative period in patients with GBM is an independent prognostic parameter that predicts progression and survival.


Subject(s)
Brain Neoplasms/surgery , Glioblastoma/surgery , Nutrition Assessment , Nutritional Status/physiology , Postoperative Care/trends , Aged , Brain Neoplasms/diagnostic imaging , Female , Glioblastoma/blood , Glioblastoma/diagnostic imaging , Humans , Lymphocytes/metabolism , Male , Middle Aged , Neutrophils/metabolism , Postoperative Care/methods , ROC Curve , Retrospective Studies
16.
Comput Biol Chem ; 92: 107458, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33761401

ABSTRACT

Glioblastoma Multiforme (GBM) is the most common, invasive, and malignant primary brain tumor with a poor prognosis and a median survival of 12-15 months. This study tried to identify the most significant miRNA biomarkers in both tissue and serum samples of GBM. GSE25632 was employed from gene expression omnibus and using WGCNA package, association of miRNA networks and clinical data was explored and brown and green modules identified as the most relevant modules. Independently, Limma package was utilized to identify differentially expressed miRNAs (DEMs) in GSE25632 by cutoff logFC > 2 and P.value < 0.05. By merging the results of Limma and WGCNA, the miRNAs that were in brown and green modules and had mentioned cutoff were selected as hub miRNAs. Performing enrichment analysis, Pathways in cancer, Prostate cancer, Glioma, p53 signaling pathway, and Focal adhesion were identified as the most important signaling pathways. Based on miRNA- target genes, has-mir-330-3p and has-mir-485-5p were identified as core miRNAs. The expression level of core miRNAs was validated by GSE90604, GSE42657, and GSE93850. We evaluated the expression level of common target genes of two detected core genes based on GSE77043, GSE42656, GSE22891, GSE15824, and GSE122498. The ability of detected miRNAs to discriminate GBM from healthy controls was assessed by area under the curve (AUC) using the ROC curve analysis. Based on TCGA database, we tested the prognostic significance of miRNAs using overall survival analysis. We evaluated the expression level of the miRNAs in tissue of 83 GBM patients and also non-tumoral adjacent (as control) tissues. We used serum samples of 34 GBM patients to evaluate the expression levels of the hub miRNAs compare to the controls. Our results showed that has-mir-330-3p and has-mir-485-5p could be potential biomarkers in GBM.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Computational Biology , Glioblastoma/genetics , MicroRNAs/genetics , Biomarkers, Tumor/blood , Brain Neoplasms/blood , Glioblastoma/blood , Humans , MicroRNAs/blood , ROC Curve
17.
Cells ; 10(3)2021 02 28.
Article in English | MEDLINE | ID: mdl-33670924

ABSTRACT

Glioblastoma (GBM) is the most common primary central nervous system tumor and one of the most lethal cancers worldwide, with morbidity of 5.26 per 100,000 population per year. These tumors are often associated with poor prognosis and terrible quality of life. Extracellular vesicles (EVs) are membrane-bound nanoparticles secreted by cells and contain lipid, protein, DNA, mRNA, miRNA and other bioactive substances. EVs perform biological functions by binding or horizontal transfer of bioactive substances to target cell receptors. In recent years, EVs have been considered as possible targets for GBM therapy. A great many types of research demonstrated that EVs played a vital role in the GBM microenvironment, development, progression, angiogenesis, invasion, and even the diagnosis of GBM. Nevertheless, the exact molecular mechanisms and roles of EVs in these processes are unclear. It can provide the basis for GBM treatment in the future that clarifying the regulatory mechanism and related signal pathways of EVs derived from GBM and their clinical value in GBM diagnosis and treatment. In this paper, the research progress and clinical application prospects of GBM-derived EVs are reviewed and discussed.


Subject(s)
Brain Neoplasms/blood , Extracellular Vesicles/metabolism , Glioblastoma/blood , Humans , Tumor Microenvironment
18.
BMC Cancer ; 21(1): 285, 2021 Mar 16.
Article in English | MEDLINE | ID: mdl-33726710

ABSTRACT

BACKGROUND: Differentiating true glioblastoma multiforme (GBM) from pseudoprogression (PsP) remains a challenge with current standard magnetic resonance imaging (MRI). The objective of this study was to explore whether patients' absolute lymphocyte count (ALC) levels can be utilized to predict true tumor progression and PsP. METHODS: Patients were considered eligible for the study if they had 1) GBM diagnosis, 2) a series of blood cell counts and clinical follow-ups, and 3) tumor progression documented by both MRI and pathology. Data analysis results include descriptive statistics, median (IQR) for continuous variables and count (%) for categorical variables, p values from Wilcoxon rank sum test or Fisher's exact test for comparison, respectively, and Kaplan-Meier analysis for overall survival (OS). OS was defined as the time from patients' second surgery to their time of death or last follow up if patients were still alive. RESULTS: 78 patients were included in this study. The median age was 56 years. Median ALC dropped 34.5% from baseline 1400 cells/mm3 to 917 cells/mm3 after completion of radiation therapy (RT) and temozolomide (TMZ). All study patients had undergone surgical biopsy upon MRI-documented progression. 37 had true tumor progression (47.44%) and 41 had pseudoprogression (52.56%). ALC before RT/TMZ, post RT/TMZ and at the time of MRI-documented progression did not show significant difference between patients with true progression and PsP. Although not statistically significant, this study found that patients with true progression had worse OS compared to those with PsP (Hazard Ratio [HR] 1.44, 95% CI 0.86-2.43, P = 0.178). This study also found that patients with high ALC (dichotomized by median) post-radiation had longer OS. CONCLUSION: Our results indicate that ALC level in GBM patients before or after treatment does not have predictive value for true disease progression or pseudoprogression. Patients with true progression had worse OS compared to those who had pseudoprogression. A larger sample size that includes CD4 cell counts may be needed to evaluate the PsP predictive value of peripheral blood biomarkers.


Subject(s)
Brain Neoplasms/diagnosis , Glioblastoma/diagnosis , Lymphocytes , Adult , Aged , Brain/diagnostic imaging , Brain/pathology , Brain Neoplasms/blood , Brain Neoplasms/mortality , Brain Neoplasms/therapy , Chemoradiotherapy/methods , Diagnosis, Differential , Disease Progression , Female , Follow-Up Studies , Glioblastoma/blood , Glioblastoma/mortality , Glioblastoma/therapy , Humans , Kaplan-Meier Estimate , Lymphocyte Count , Magnetic Resonance Imaging , Male , Middle Aged , Predictive Value of Tests , Progression-Free Survival , Retrospective Studies , Temozolomide/therapeutic use
19.
World Neurosurg ; 149: e758-e765, 2021 05.
Article in English | MEDLINE | ID: mdl-33540096

ABSTRACT

BACKGROUND: Some patients with glioblastoma multiforme (GBM) survive 3-5 years (or longer) after diagnosis. The goal of this study was to identify differences between the long-term survivors (LTS) and those who had a shorter overall survival (non-LTS groups). METHODS: This study was a retrospective analysis of prospectively maintained surgical databases. All patients who underwent safe maximal resection for GBM were included. Demographic, clinical, radiologic, and pathologic data were obtained from electronic medical records. Values of the biomarkers of systemic inflammation were computed from the preoperative hemogram reports. Patients with an overall survival (OS) ≥36 months were defined as the LTS group and were compared with the non-LTS groups (OS<36 months). RESULTS: Patients in the LTS group were younger, had a better baseline performance status, and were more likely to have undergone near- or gross-total resection. LTS was associated with lower Ki67 labeling, MGMT methylation, IDH mutation, and lack of p53 overexpression. Several novel findings were generated by this study. A longer pretreatment duration of symptoms was associated with a longer OS. Higher pretreatment levels of the absolute neutrophil count, neutrophil-lymphocyte ratio, monocyte-lymphocyte ratio, derived neutrophil-lymphocyte ratio and systemic index of inflammation, and lower levels of the absolute eosinophil count and eosinophil-lymphocyte ratio all correlated with a shorter OS. CONCLUSIONS: Several differences were identified between the LTS and non-LTS groups. These differences will likely be incorporated into future prognostic models. They may also aid in differentiation between recurrent disease and treatment-related changes.


Subject(s)
Brain Neoplasms/surgery , Cancer Survivors , Glioblastoma/surgery , Adult , Age Factors , Brain Neoplasms/blood , Brain Neoplasms/genetics , Brain Neoplasms/physiopathology , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Eosinophils , Female , Glioblastoma/blood , Glioblastoma/genetics , Glioblastoma/physiopathology , Humans , Isocitrate Dehydrogenase/genetics , Karnofsky Performance Status , Ki-67 Antigen/metabolism , Leukocyte Count , Lymphocyte Count , Male , Margins of Excision , Middle Aged , Monocytes , Mutation , Neutrophils , Platelet Count , Prognosis , Retrospective Studies , Survival Rate , Time Factors , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics
20.
Oncogene ; 40(8): 1516-1530, 2021 02.
Article in English | MEDLINE | ID: mdl-33452462

ABSTRACT

The basic leucine zipper ATF-like transcription factor 2 (BATF2) has been implicated in inflammatory responses and anti-tumour effects. Little, however, is known regarding its extracellular role in maintaining a non-supportive cancer microenvironment. Here, we show that BATF2 inhibits glioma growth and myeloid-derived suppressor cells (MDSCs) recruitment. Interestingly, extracellular vesicles (EVs) from BATF2-overexpressing glioma cell lines (BATF2-EVs) inhibited MDSCs chemotaxis in vitro. Moreover, BATF2 inhibited intracellular SDF-1α and contributes to decreased SDF-1α in EVs. In addition, BATF2 downregulation-induced MDSCs recruitment were reversed by blocking SDF-1α/CXCR4 signalling upon AMD3100 treatment. Specifically, detection of EVs in 24 pairs of gliomas and healthy donors at different stages revealed that the abundance of BATF2-positive EVs in plasma (BATF2+ plEVs) can distinguish stage III-IV glioma from stage I-II glioma and healthy donors. Taken together, our study identified novel regulatory functions of BATF2 in regulating MDSCs recruitment, providing a prognostic value in terms of the number of BATF2+ plEVs in glioma stage.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Chemokine CXCL12/genetics , Glioblastoma/genetics , Receptors, CXCR4/genetics , Tumor Suppressor Proteins/genetics , Adult , Aged , Animals , Benzylamines/pharmacology , Cell Line, Tumor , Cell Migration Inhibition/genetics , Cyclams/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/blood , Glioblastoma/pathology , Heterografts , Humans , Male , Mice , Middle Aged , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , Neoplasm Staging , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...