Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
NMR Biomed ; 37(4): e5095, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38213096

ABSTRACT

The standard treatment in glioblastoma includes maximal safe resection followed by concomitant radiotherapy plus chemotherapy and adjuvant temozolomide. The first follow-up study to evaluate treatment response is performed 1 month after concomitant treatment, when contrast-enhancing regions may appear that can correspond to true progression or pseudoprogression. We retrospectively evaluated 31 consecutive patients at the first follow-up after concomitant treatment to check whether the metabolic pattern assessed with multivoxel MRS was predictive of treatment response 2 months later. We extracted the underlying metabolic patterns of the contrast-enhancing regions with a blind-source separation method and mapped them over the reference images. Pattern heterogeneity was calculated using entropy, and association between patterns and outcomes was measured with Cramér's V. We identified three distinct metabolic patterns-proliferative, necrotic, and responsive, which were associated with status 2 months later. Individually, 70% of the patients showed metabolically heterogeneous patterns in the contrast-enhancing regions. Metabolic heterogeneity was not related to the regions' size and only stable patients were less heterogeneous than the rest. Contrast-enhancing regions are also metabolically heterogeneous 1 month after concomitant treatment. This could explain the reported difficulty in finding robust pseudoprogression biomarkers.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/therapy , Glioblastoma/drug therapy , Follow-Up Studies , Retrospective Studies , Dacarbazine/therapeutic use , Chemoradiotherapy/methods , Disease Progression , Brain Neoplasms/therapy , Brain Neoplasms/drug therapy , Magnetic Resonance Imaging/methods
2.
Int J Mol Sci ; 24(24)2023 Dec 18.
Article in English | MEDLINE | ID: mdl-38139457

ABSTRACT

The tumor microenvironment in glioblastoma (GB) is considered to be "cold", i.e., the fraction of cytotoxic T cells, for instance, is low. Instead, macrophages are the major immune cell population in GB, which stem either from tissue response (resident microglia) or recruitment of macrophages from the periphery, thereby undergoing tumor-dependent "imprinting" mechanisms by which macrophages can adapt a tumor-supportive phenotype. In this regard, it is important to describe the nature of macrophages associated with GB, in particular under therapy conditions using the gold standard chemotherapy drug temozolomide (TMZ). Here, we explored the suitability of combining information from in vivo magnetic resonance spectroscopic (MRS) approaches (metabolomics) with in vitro molecular analyses to assess therapy response and characterize macrophage populations in mouse GB using an isogenic GL261 model. For macrophage profiling, expression levels of matrix metalloproteinases (MMPs) and A disintegrin and metalloproteinases (ADAMs) were determined, since their gene products affect macrophage-tumor cell communication by extensive cleavage of immunomodulatory membrane proteins, such as PD-L1. In tumor mice with an overall therapy response, expression of genes encoding the proteases ADAM8, ADAM10, and ADAM17 was increased and might contribute to the immunosuppressive phenotype of GB and immune cells. In tumors responding to therapy, expression levels of ADAM8 were upregulated by TMZ, and higher levels of PD-L1 were correlated significantly. Using a CRISPR/Cas9 knockout of ADAM8 in GL261 cells, we demonstrated that soluble PD-L1 (sPD-L1) is only generated in the presence of ADAM8. Moreover, primary macrophages from WT and ADAM8-deficient mice showed ADAM8-dependent release of sPD-L1, independent of the macrophage polarization state. Since ADAM8 expression is induced in responding tumors and PD-L1 shedding is likely to decrease the anti-tumor activities of T-cells, we conclude that immunotherapy resistance is caused, at least in part, by the increased presence of proteases, such as ADAM8.


Subject(s)
Glioblastoma , Glioma , Animals , Mice , Temozolomide/pharmacology , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , B7-H1 Antigen/metabolism , Tumor Microenvironment/genetics , Glioma/pathology , Cell Line, Tumor
3.
NMR Biomed ; 36(12): e5020, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37582395

ABSTRACT

Magnetic resonance spectroscopy (MRS) is an MR technique that provides information about the biochemistry of tissues in a noninvasive way. MRS has been widely used for the study of brain tumors, both preoperatively and during follow-up. In this study, we investigated the performance of a range of variants of unsupervised matrix factorization methods of the non-negative matrix underapproximation (NMU) family, namely, sparse NMU, global NMU, and recursive NMU, and compared them with convex non-negative matrix factorization (C-NMF), which has previously shown a good performance on brain tumor diagnostic support problems using MRS data. The purpose of the investigation was 2-fold: first, to ascertain the differences among the sources extracted by these methods; and second, to compare the influence of each method in the diagnostic accuracy of the classification of brain tumors, using them as feature extractors. We discovered that, first, NMU variants found meaningful sources in terms of biological interpretability, but representing parts of the spectrum, in contrast to C-NMF; and second, that NMU methods achieved better classification accuracy than C-NMF for the classification tasks when one class was not meningioma.


Subject(s)
Brain Neoplasms , Meningeal Neoplasms , Meningioma , Humans , Brain Neoplasms/pathology , Magnetic Resonance Spectroscopy/methods , Algorithms
4.
Cancers (Basel) ; 15(14)2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37509372

ABSTRACT

In vivo magnetic resonance spectroscopy (MRS) has two modalities, single-voxel (SV) and multivoxel (MV), in which one or more contiguous grids of SVs are acquired. PURPOSE: To test whether MV grids can be classified with models trained with SV. METHODS: Retrospective study. Training dataset: Multicenter multiformat SV INTERPRET, 1.5T. Testing dataset: MV eTumour, 3T. Two classification tasks were completed: 3-class (meningioma vs. aggressive vs. normal) and 4-class (meningioma vs. low-grade glioma vs. aggressive vs. normal). Five different methods were tested for feature selection. The classification was implemented using linear discriminant analysis (LDA), random forest, and support vector machines. The evaluation was completed with balanced error rate (BER) and area under the curve (AUC) on both sets. The accuracy in class prediction was calculated by developing a solid tumor index (STI) and segmentation accuracy with the Dice score. RESULTS: The best method was sequential forward feature selection combined with LDA, with AUCs = 0.95 (meningioma), 0.89 (aggressive), 0.82 (low-grade glioma), and 0.82 (normal). STI was 66% (4-class task) and 71% (3-class task) because two cases failed completely and two more had suboptimal STI as defined by us. DISCUSSION: The reasons for failure in the classification of the MV test set were related to the presence of artifacts.

5.
Nanomaterials (Basel) ; 13(10)2023 May 12.
Article in English | MEDLINE | ID: mdl-37242036

ABSTRACT

Current therapies for treating Glioblastoma (GB), and brain tumours in general, are inefficient and represent numerous challenges. In addition to surgical resection, chemotherapy and radiotherapy are presently used as standards of care. However, treated patients still face a dismal prognosis with a median survival below 15-18 months. Temozolomide (TMZ) is the main chemotherapeutic agent administered; however, intrinsic or acquired resistance to TMZ contributes to the limited efficacy of this drug. To circumvent the current drawbacks in GB treatment, a large number of classical and non-classical platinum complexes have been prepared and tested for anticancer activity, especially platinum (IV)-based prodrugs. Platinum complexes, used as alkylating agents in the anticancer chemotherapy of some malignancies, are though often associated with severe systemic toxicity (i.e., neurotoxicity), especially after long-term treatments. The objective of the current developments is to produce novel nanoformulations with improved lipophilicity and passive diffusion, promoting intracellular accumulation, while reducing toxicity and optimizing the concomitant treatment of chemo-/radiotherapy. Moreover, the blood-brain barrier (BBB) prevents the access of the drugs to the brain and accumulation in tumour cells, so it represents a key challenge for GB management. The development of novel nanomedicines with the ability to (i) encapsulate Pt-based drugs and pro-drugs, (ii) cross the BBB, and (iii) specifically target cancer cells represents a promising approach to increase the therapeutic effect of the anticancer drugs and reduce undesired side effects. In this review, a critical discussion is presented concerning different families of nanoparticles able to encapsulate platinum anticancer drugs and their application for GB treatment, emphasizing their potential for increasing the effectiveness of platinum-based drugs.

6.
Metabolites ; 12(3)2022 Mar 14.
Article in English | MEDLINE | ID: mdl-35323686

ABSTRACT

This hypothesis proposal addresses three major questions: (1) Why do we need imaging biomarkers for assessing the efficacy of immune system participation in glioblastoma therapy response? (2) Why are they not available yet? and (3) How can we produce them? We summarize the literature data supporting the claim that the immune system is behind the efficacy of most successful glioblastoma therapies but, unfortunately, there are no current short-term imaging biomarkers of its activity. We also discuss how using an immunocompetent murine model of glioblastoma, allowing the cure of mice and the generation of immune memory, provides a suitable framework for glioblastoma therapy response biomarker studies. Both magnetic resonance imaging and magnetic resonance-based metabolomic data (i.e., magnetic resonance spectroscopic imaging) can provide non-invasive assessments of such a system. A predictor based in nosological images, generated from magnetic resonance spectroscopic imaging analyses and their oscillatory patterns, should be translational to clinics. We also review hurdles that may explain why such an oscillatory biomarker was not reported in previous imaging glioblastoma work. Single shot explorations that neglect short-term oscillatory behavior derived from immune system attack on tumors may mislead actual response extent detection. Finally, we consider improvements required to properly predict immune system-mediated early response (1-2 weeks) to therapy. The sensible use of improved biomarkers may enable translatable evidence-based therapeutic protocols, with the possibility of extending preclinical results to human patients.

7.
NMR Biomed ; 35(4): e4193, 2022 04.
Article in English | MEDLINE | ID: mdl-31793715

ABSTRACT

Despite the success of automated pattern recognition methods in problems of human brain tumor diagnostic classification, limited attention has been paid to the issue of automated data quality assessment in the field of MRS for neuro-oncology. Beyond some early attempts to address this issue, the current standard in practice is MRS quality control through human (expert-based) assessment. One aspect of automatic quality control is the problem of detecting artefacts in MRS data. Artefacts, whose variety has already been reviewed in some detail and some of which may even escape human quality control, have a negative influence in pattern recognition methods attempting to assist tumor characterization. The automatic detection of MRS artefacts should be beneficial for radiology as it guarantees more reliable tumor characterizations, as well as the development of more robust pattern recognition-based tumor classifiers and more trustable MRS data processing and analysis pipelines. Feature extraction methods have previously been used to help distinguishing between good and bad quality spectra to apply subsequent supervised pattern recognition techniques. In this study, we apply feature extraction differently and use a variant of a method for blind source separation, namely Convex Non-Negative Matrix Factorization, to unveil MRS signal sources in a completely unsupervised way. We hypothesize that, while most sources will correspond to the different tumor patterns, some of them will reflect signal artefacts. The experimental work reported in this paper, analyzing a combined short and long echo time 1 H-MRS database of more than 2000 spectra acquired at 1.5T and corresponding to different tumor types and other anomalous masses, provides a first proof of concept that points to the possible validity of this approach.


Subject(s)
Algorithms , Brain Neoplasms , Artifacts , Brain Neoplasms/pathology , Humans , Pattern Recognition, Automated/methods , Quality Control
8.
Cancers (Basel) ; 13(11)2021 May 28.
Article in English | MEDLINE | ID: mdl-34071393

ABSTRACT

Glioblastomas (GB) are brain tumours with poor prognosis even after aggressive therapy. Previous work suggests that magnetic resonance spectroscopic imaging (MRSI) could act as a biomarker of efficient immune system attack onto GB, presenting oscillatory changes. Glioma-associated microglia/macrophages (GAMs) constitute the most abundant non-tumour cell type within the GB and can be polarised into anti-tumour (M1) or pro-tumour (M2) phenotypes. One of the mechanisms to mediate immunosuppression in brain tumours is the interaction between programmed cell death-1 ligand 1 (PD-L1) and programmed cell death-1 receptor (PD-1). We evaluated the subpopulations of GAMs in responding and control GB tumours to correlate PD-L1 expression to GAM polarisation in order to explain/validate MRSI-detected findings. Mice were evaluated by MRI/MRSI to assess the extent of response to treatment and with qPCR for GAMs M1 and M2 polarisation analyses. M1/M2 ratios and PD-L1 expression were higher in treated compared to control tumours. Furthermore, PD-L1 expression was positively correlated with the M1/M2 ratio. The oscillatory change in the GAMs prevailing population could be one of the key causes for the differential MRSI-detected pattern, allowing this to act as immune system activity biomarker in future work.

9.
Int J Mol Sci ; 22(7)2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33810611

ABSTRACT

BACKGROUND: The relevance of the cancer immune cycle in therapy response implies that successful treatment may trigger the exposure or the release of immunogenic signals. Previous results with the preclinical GL261 glioblastoma (GB) showed that combination treatment of temozolomide (TMZ) + CX-4945 (protein kinase CK2 inhibitor) outperformed single treatments, provided an immune-friendly schedule was followed. Our purpose was to study possible immunogenic signals released in vitro by GB cells. METHODS: GL261 GB cells were treated with TMZ and CX-4945 at different concentrations (25 µM-4 mM) and time frames (12-72 h). Cell viability was measured with Trypan Blue and propidium iodide. Calreticulin exposure was assessed with immunofluorescence, and ATP release was measured with bioluminescence. RESULTS: TMZ showed cytostatic rather than cytotoxic effects, while CX-4945 showed remarkable cytotoxic effects already at low concentrations. Calreticulin exposure after 24 h was detected with TMZ treatment, as well as TMZ/CX-4945 low concentration combined treatment. ATP release was significantly higher with CX-4945, especially at high concentrations, as well as with TMZ/CX-4945. CONCLUSIONS: combined treatment may produce the simultaneous release of two potent immunogenic signals, which can explain the outperformance over single treatments in vivo. A word of caution may be raised since in vitro conditions are not able to mimic pharmacokinetics observed in vivo fully.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Naphthyridines/administration & dosage , Phenazines/administration & dosage , Temozolomide/administration & dosage , Adenosine Triphosphate/chemistry , Antineoplastic Agents, Alkylating/administration & dosage , Calreticulin/chemistry , Casein Kinase II/metabolism , Cell Line, Tumor , Cell Survival , Combined Modality Therapy , Humans , Inflammation , Microscopy, Fluorescence , Propidium/chemistry , Signal Transduction , Treatment Outcome
10.
Sci Rep ; 10(1): 19699, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33184423

ABSTRACT

Glioblastoma is the most frequent aggressive primary brain tumor amongst human adults. Its standard treatment involves chemotherapy, for which the drug temozolomide is a common choice. These are heterogeneous and variable tumors which might benefit from personalized, data-based therapy strategies, and for which there is room for improvement in therapy response follow-up, investigated with preclinical models. This study addresses a preclinical question that involves distinguishing between treated and control (untreated) mice bearing glioblastoma, using machine learning techniques, from magnetic resonance-based data in two modalities: MRI and MRSI. It aims to go beyond the comparison of methods for such discrimination to provide an analytical pipeline that could be used in subsequent human studies. This analytical pipeline is meant to be a usable and interpretable tool for the radiology expert in the hope that such interpretation helps revealing new insights about the problem itself. For that, we propose coupling source extraction-based and radiomics-based data transformations with feature selection. Special attention is paid to the generation of radiologist-friendly visual nosological representations of the analyzed tumors.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Pattern Recognition, Automated/methods , Temozolomide/administration & dosage , Animals , Brain Neoplasms/diagnostic imaging , Cell Line, Tumor , Glioblastoma/diagnostic imaging , Humans , Machine Learning , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Mice , Retrospective Studies , Temozolomide/therapeutic use , Treatment Outcome , Xenograft Model Antitumor Assays
11.
Int J Mol Sci ; 21(22)2020 Nov 20.
Article in English | MEDLINE | ID: mdl-33233585

ABSTRACT

Glioblastomas (GBs) are malignant brain tumours with poor prognosis even after aggressive therapy. Programmed cell death-1 (PD-1) immune checkpoint blockade is a promising strategy in many types of cancer, but its therapeutic effects in GB remain low and associated with immune infiltration. Previous work suggests that oscillations of magnetic resonance spectroscopic imaging (MRSI)-based response pattern with chemotherapy could act as a biomarker of efficient immune system attack onto GBs. The presence of such oscillations with other monotherapies such as anti-PD-1 would reinforce its monitoring potential. Here, we confirm that the oscillatory behaviour of the response biomarker is also detected in mice treated with anti PD-1 immunotherapy both in combination with temozolomide and as monotherapy. This indicates that the spectral pattern changes observed during therapy response are shared by different therapeutic strategies, provided the host immune system is elicited and able to productively attack tumour cells. Moreover, the participation of the immune system in response is also supported by the rate of cured animals observed with different therapeutic strategies (in the range of 50-100% depending on the treatment), which also held long-term immune memory against tumour cells re-challenge. Taken together, our findings open the way for a translational use of the MRSI-based biomarker in patient-tailored GB therapy, including immunotherapy, for which reliable non-invasive biomarkers are still missing.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Programmed Cell Death 1 Receptor/genetics , Temozolomide/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biomarkers, Pharmacological , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Drug Administration Schedule , Drug Chronotherapy , Drug Evaluation, Preclinical , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/diagnostic imaging , Glioblastoma/genetics , Glioblastoma/mortality , Immunoglobulin G/pharmacology , Immunologic Memory/drug effects , Immunotherapy/methods , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Survival Analysis , Tumor Burden/drug effects
12.
NMR Biomed ; 33(4): e4229, 2020 04.
Article in English | MEDLINE | ID: mdl-31926117

ABSTRACT

Glioblastomas (GB) are brain tumours with poor prognosis even after aggressive therapy. Improvements in both therapeutic and follow-up strategies are urgently needed. In previous work we described an oscillatory pattern of response to Temozolomide (TMZ) using a standard administration protocol, detected through MRSI-based machine learning approaches. In the present work, we have introduced the Immune-Enhancing Metronomic Schedule (IMS) with an every 6-d TMZ administration at 60 mg/kg and investigated the consistence of such oscillatory behaviour. A total of n = 17 GL261 GB tumour-bearing C57BL/6j mice were studied with MRI/MRSI every 2 d, and the oscillatory behaviour (6.2 ± 1.5 d period from the TMZ administration day) was confirmed during response. Furthermore, IMS-TMZ produced significant improvement in mice survival (22.5 ± 3.0 d for controls vs 135.8 ± 78.2 for TMZ-treated), outperforming standard TMZ treatment. Histopathological correlation was investigated in selected tumour samples (n = 6) analyzing control and responding fields. Significant differences were found for CD3+ cells (lymphocytes, 3.3 ± 2.5 vs 4.8 ± 2.9, respectively) and Iba-1 immunostained area (microglia/macrophages, 16.8% ± 9.7% and 21.9% ± 11.4%, respectively). Unexpectedly, during IMS-TMZ treatment, tumours from some mice (n = 6) fully regressed and remained undetectable without further treatment for 1 mo. These animals were considered "cured" and a GL261 re-challenge experiment performed, with no tumour reappearance in five out of six cases. Heterogeneous therapy response outcomes were detected in tumour-bearing mice, and a selected group was investigated (n = 3 non-responders, n = 6 relapsing tumours, n = 3 controls). PD-L1 content was found ca. 3-fold increased in the relapsing group when comparing with control and non-responding groups, suggesting that increased lymphocyte inhibition could be associated to IMS-TMZ failure. Overall, data suggest that host immune response has a relevant role in therapy response/escape in GL261 tumours under IMS-TMZ therapy. This is associated to changes in the metabolomics pattern, oscillating every 6 d, in agreement with immune cycle length, which is being sampled by MRSI-derived nosological images.


Subject(s)
Administration, Metronomic , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/immunology , Magnetic Resonance Imaging , Temozolomide/administration & dosage , Temozolomide/therapeutic use , Animals , B7-H1 Antigen/metabolism , Cell Line, Tumor , Glioblastoma/diagnostic imaging , Glioblastoma/pathology , Humans , Immunologic Memory/drug effects , Mice, Inbred C57BL , Tumor Burden/drug effects
13.
J Neuropathol Exp Neurol ; 79(2): 184-193, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31846038

ABSTRACT

Dogs develop gliomas with similar histopathological features to human gliomas and share with them the limited success of current therapeutic regimens such as surgery and radiation. The tumor microenvironment in gliomas is influenced by immune cell infiltrates. The present study aims to immunohistochemically characterize the tumor-infiltrating lymphocyte (TIL) population of naturally occurring canine gliomas, focusing on the expression of Forkhead box P3-positive (FOXP3+) regulatory T-cells (Tregs). Forty-three canine gliomas were evaluated immunohistochemically for the presence of CD3+, FOXP3+, and CD20+ TILs. In low-grade gliomas, CD3+ TILs were found exclusively within the tumor tissue. In high-grade gliomas, they were present in significantly higher numbers throughout the tumor and in the brain-tumor junction. CD20+ TILs were rarely found in comparison to CD3+ TILs. FOXP3+ TILs shared a similar distribution with CD3+ TILs. The accumulation of FOXP3+ Tregs within the tumor was more pronounced in astrocytic gliomas than in tumors of oligodendroglial lineage and the difference in expression was significant when comparing low-grade oligodendrogliomas and high-grade astrocytomas. Only high-grade astrocytomas presented FOXP3+ cells with tumoral morphology. In spontaneous canine gliomas, TILs display similar characteristics (density and distribution) as described for human gliomas, supporting the use of the dog as an animal model for translational immunotherapeutic studies.


Subject(s)
Brain Neoplasms/metabolism , Forkhead Transcription Factors/metabolism , Glioma/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Animals , Antigens, CD20/metabolism , Brain Neoplasms/complications , Brain Neoplasms/pathology , CD3 Complex/metabolism , Dogs , Female , Glioma/complications , Glioma/pathology , Lymphocytes, Tumor-Infiltrating/pathology , Male
14.
Pathol Oncol Res ; 25(4): 1659-1663, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30607803

ABSTRACT

Glioblastoma (GB) is the most prevalent malignant primary brain tumor in adults. The preclinical glioblastoma model GL261 is widely used for investigating new therapeutic strategies. GL261 cultured cells are used for assessing preliminary in vitro data for this model although very little is known about the molecular characteristics of this cell line. Protein Kinase CK2 is a pleiotropic serine-threonine kinase and its inhibition may be a promising therapeutic strategy for GB treatment. In our group we follow treatment response with CK2 inhibitors in vivo using the GL261 murine model. For that, it is of our interest to assess the differential expression of α, α', ß CK2 subunits as well as CK2 activity in the GL261 GB model. CK2α' expression changed along the growth curve of GL261 cells, undergoing downregulation in postconfluent phase cells, whereas CK2α and CK2ß expression remained essentially unchanged. Furthermore, a marked decrease in CK2 activity in slowly proliferating postconfluent phase GL261 cells was observed. Finally, CK2α' expression in orthotopic GL261 tumors was intermediate between CK2α' expression found in cultured cells in exponentially growing or postconfluent phase, reflecting the heterogeneous nature of GL261 tumours growing in vivo. The results obtained suggest that, in the GL261 cell line, CK2α' could play a specific role in highly proliferative cells. Also, the decrease in CK2 activity in slowly proliferating GL261 cells could imply a differential susceptibility to subunit-specific CK2 inhibitors in this cell line, although further studies are needed to confirm this hypothesis.


Subject(s)
Biomarkers, Tumor/metabolism , Casein Kinase II/metabolism , Cell Proliferation , Disease Models, Animal , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Animals , Glioblastoma/enzymology , Mice , Tumor Cells, Cultured , Up-Regulation
15.
NMR Biomed ; 32(10): e4054, 2019 10.
Article in English | MEDLINE | ID: mdl-30633389

ABSTRACT

The contribution of MRS(I) to the in vivo evaluation of cancer-metabolism-derived metrics, mostly since 2016, is reviewed here. Increased carbon consumption by tumour cells, which are highly glycolytic, is now being sampled by 13 C magnetic resonance spectroscopic imaging (MRSI) following the injection of hyperpolarized [1-13 C] pyruvate (Pyr). Hot-spots of, mostly, increased lactate dehydrogenase activity or flow between Pyr and lactate (Lac) have been seen with cancer progression in prostate (preclinical and in humans), brain and pancreas (both preclinical) tumours. Therapy response is usually signalled by decreased Lac/Pyr 13 C-labelled ratio with respect to untreated or non-responding tumour. For therapeutic agents inducing tumour hypoxia, the 13 C-labelled Lac/bicarbonate ratio may be a better metric than the Lac/Pyr ratio. 31 P MRSI may sample intracellular pH changes from brain tumours (acidification upon antiangiogenic treatment, basification at fast proliferation and relapse). The steady state tumour metabolome pattern is still in use for cancer evaluation. Metrics used for this range from quantification of single oncometabolites (such as 2-hydroxyglutarate in mutant IDH1 glial brain tumours) to selected metabolite ratios (such as total choline to N-acetylaspartate (plain ratio or CNI index)) or the whole 1 H MRSI(I) pattern through pattern recognition analysis. These approaches have been applied to address different questions such as tumour subtype definition, following/predicting the response to therapy or defining better resection or radiosurgery limits.


Subject(s)
Magnetic Resonance Spectroscopy , Neoplasms/metabolism , Neoplasms/pathology , Animals , Choline/metabolism , Humans , Hydrogen-Ion Concentration , Metabolome , Neoplasms/diagnostic imaging , Neoplasms/therapy , Succinic Acid/metabolism
16.
Eur Radiol ; 29(6): 2792-2801, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30569184

ABSTRACT

OBJECTIVES: Assessing a posterior fossa tumour in an adult can be challenging. Metastasis, haemangioblastoma, ependymal tumours, and medulloblastoma are the most common diagnostic possibilities. Our aim was to evaluate the contribution of magnetic resonance spectroscopy (MRS) in the diagnosis of these entities. METHODS: We retrospectively evaluated 56 consecutive patients with a posterior fossa tumour and histological diagnosis of ependymal tumour, medulloblastoma, haemangioblastoma, and metastasis in which good-quality spectra at short (TE 30 ms) or/and intermediate (TE, 136 ms) TE were available. Spectra were compared using the Mann-Whitney U non-parametric test in order to select the spectral datapoints and the intensity ratios that showed significant differences between groups of lesions. Performance of these datapoints and their ratios were assessed with ROC curves. RESULTS: The most characteristic signatures on spectroscopy were high choline (Cho) in medulloblastoma (p < 0.001), high myoinositol (mIns) in ependymal tumours (p < 0.05), and high lipids (LIP) in haemangioblastoma (p < 0.01) and metastasis (p < 0.01). Selected ratios between normalised intensity signals of resonances provided accuracy values between 79 and 95% for pairwise comparisons. Intensity ratio NI3.21ppm/3.55ppm provided satisfactory discrimination between medulloblastoma and ependymal tumours (accuracy, 92%), ratio NI2.11ppm/1.10ppm discriminated ependymal tumours from haemangioblastoma (accuracy, 94%), ratio NI3.21ppm/1.13ppm discriminated haemangioblastoma from medulloblastoma (accuracy, 95%), and ratio NI1.28ppm/2.02pmm discriminated haemangioblastoma from metastasis (accuracy, 83%). CONCLUSIONS: MRS may improve the non-invasive diagnosis of posterior fossa tumours in adults. KEY POINTS: • High choline suggests a medulloblastoma in a posterior fossa tumour. • High myoinositol suggests an ependymal lesion in a posterior fossa tumour. • High lipids suggest a metastasis or a haemangioblastoma in a posterior fossa tumour.


Subject(s)
Choline/metabolism , Hemangioblastoma/diagnosis , Infratentorial Neoplasms/diagnosis , Inositol/metabolism , Magnetic Resonance Spectroscopy/methods , Medulloblastoma/diagnosis , Adult , Biomarkers, Tumor/metabolism , Diagnosis, Differential , Female , Hemangioblastoma/metabolism , Hemangioblastoma/secondary , Humans , Magnetic Resonance Imaging/methods , Male , Medulloblastoma/metabolism , Medulloblastoma/secondary , Neoplasm Metastasis , ROC Curve , Retrospective Studies , Young Adult
17.
J Comp Neurol ; 526(4): 721-741, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29205371

ABSTRACT

The mammalian ventricular-subventricular zone (V-SVZ) presents the highest neurogenic potential in the brain of the adult individual. In rodents, it is mainly composed of chains of neuroblasts. In humans, it is organized in layers where neuroblasts do not form chains. The aim of this study is to describe the cytoarchitecture of canine V-SVZ (cV-SVZ), to assess its neurogenic potential, and to compare our results with those previously described in other species. We have studied by histology, immunohistochemistry (IHC), electron microscopy and neurosphere assay the morphology, cytoarchitecture and neurogenic potential of cV-SVZ. Age groups of animals were performed. Histological and ultrastructural studies indicated that the cV-SVZ is organized in layers as in humans, but including migratory chains as in rodents. Neural progenitors were organized in niches in the subependymal area and a decline in their number was observed with age. Adult-young dogs contained migratory cells capable to expand and differentiate in vitro according with previous results obtained in rodents, primates, humans, pigs, and dogs. Some adult animals presented perivascular niches outside the V-SVZ. Our observations evidence a great similarity between canine and human V-SVZ indicating that the dog may be better representative of neurogenic events in humans, compared with rodents. Accordingly with our results, we conclude that dogs are a valuable animal model of adult neurogenesis in comparative and preclinical studies.


Subject(s)
Brain/cytology , Brain/metabolism , Dogs/anatomy & histology , Dogs/metabolism , Stem Cell Niche , Animals , Brain/growth & development , Brain/ultrastructure , Cells, Cultured , Dogs/growth & development , Female , Immunohistochemistry , Male , Microscopy, Electron , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/ultrastructure , Species Specificity
18.
Magn Reson Med ; 79(5): 2500-2510, 2018 05.
Article in English | MEDLINE | ID: mdl-28994492

ABSTRACT

PURPOSE: To investigate and compare human judgment and machine learning tools for quality assessment of clinical MR spectra of brain tumors. METHODS: A very large set of 2574 single voxel spectra with short and long echo time from the eTUMOUR and INTERPRET databases were used for this analysis. Original human quality ratings from these studies as well as new human guidelines were used to train different machine learning algorithms for automatic quality control (AQC) based on various feature extraction methods and classification tools. The performance was compared with variance in human judgment. RESULTS: AQC built using the RUSBoost classifier that combats imbalanced training data performed best. When furnished with a large range of spectral and derived features where the most crucial ones had been selected by the TreeBagger algorithm it showed better specificity (98%) in judging spectra from an independent test-set than previously published methods. Optimal performance was reached with a virtual three-class ranking system. CONCLUSION: Our results suggest that feature space should be relatively large for the case of MR tumor spectra and that three-class labels may be beneficial for AQC. The best AQC algorithm showed a performance in rejecting spectra that was comparable to that of a panel of human expert spectroscopists. Magn Reson Med 79:2500-2510, 2018. © 2017 International Society for Magnetic Resonance in Medicine.


Subject(s)
Brain Neoplasms/diagnostic imaging , Image Interpretation, Computer-Assisted/methods , Machine Learning , Magnetic Resonance Imaging/methods , Algorithms , Brain/diagnostic imaging , Humans , Quality Control
19.
NMR Biomed ; 30(9)2017 Sep.
Article in English | MEDLINE | ID: mdl-28570014

ABSTRACT

Glioblastoma (GBM) causes poor survival in patients even when applying aggressive treatment. Temozolomide (TMZ) is the standard chemotherapeutic choice for GBM treatment, but resistance always ensues. In previous years, efforts have focused on new therapeutic regimens with conventional drugs to activate immune responses that may enhance tumor regression and prevent regrowth, for example the "metronomic" approaches. In metronomic scheduling studies, cyclophosphamide (CPA) in GL261 GBM growing subcutaneously in C57BL/6 mice was shown not only to activate antitumor CD8+ T-cell response, but also to induce long-term specific T-cell tumor memory. Accordingly, we have evaluated whether metronomic CPA or TMZ administration could increase survival in orthotopic GL261 in C57BL/6 mice, an immunocompetent model. Longitudinal in vivo studies with CPA (140 mg/kg) or TMZ (range 140-240 mg/kg) metronomic administration (every 6 days) were performed in tumor-bearing mice. Tumor evolution was monitored at 7 T with MRI (T2 -weighted, diffusion-weighted imaging) and MRSI-based nosological images of response to therapy. Obtained results demonstrated that both treatments resulted in increased survival (38.6 ± 21.0 days, n = 30) compared with control (19.4 ± 2.4 days, n = 18). Best results were obtained with 140 mg/kg TMZ (treated, 44.9 ± 29.0 days, n = 12, versus control, 19.3 ± 2.3 days, n = 12), achieving a longer survival rate than previous group work using three cycles of TMZ therapy at 60 mg/kg (33.9 ± 11.7 days, n = 38). Additional interesting findings were, first, clear edema appearance during chemotherapeutic treatment, second, the ability to apply the semi-supervised source analysis previously developed in our group for non-invasive TMZ therapy response monitoring to detect CPA-induced response, and third, the necropsy findings in mice cured from GBM after high TMZ cumulative dosage (980-1400 mg/kg), which demonstrated lymphoma incidence. In summary, every 6 day administration schedule of TMZ or CPA improves survival in orthotopic GL261 GBM with respect to controls or non-metronomic therapy, in partial agreement with previous work on subcutaneous GL261.


Subject(s)
Brain Neoplasms/drug therapy , Cyclophosphamide/administration & dosage , Cyclophosphamide/therapeutic use , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Immunocompetence , Administration, Metronomic , Animals , Brain Neoplasms/pathology , Cause of Death , Cell Line, Tumor , Cyclophosphamide/pharmacology , Dacarbazine/administration & dosage , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Diffusion , Female , Glioblastoma/pathology , Magnetic Resonance Imaging , Mice, Inbred C57BL , Temozolomide , Treatment Outcome , Tumor Burden/drug effects
20.
Metabolites ; 7(2)2017 May 18.
Article in English | MEDLINE | ID: mdl-28524099

ABSTRACT

Glioblastoma (GBM) is the most common aggressive primary brain tumor in adults, with a short survival time even after aggressive therapy. Non-invasive surrogate biomarkers of therapy response may be relevant for improving patient survival. Previous work produced such biomarkers in preclinical GBM using semi-supervised source extraction and single-slice Magnetic Resonance Spectroscopic Imaging (MRSI). Nevertheless, GBMs are heterogeneous and single-slice studies could prevent obtaining relevant information. The purpose of this work was to evaluate whether a multi-slice MRSI approach, acquiring consecutive grids across the tumor, is feasible for preclinical models and may produce additional insight into therapy response. Nosological images were analyzed pixel-by-pixel and a relative responding volume, the Tumor Responding Index (TRI), was defined to quantify response. Heterogeneous response levels were observed and treated animals were ascribed to three arbitrary predefined groups: high response (HR, n = 2), TRI = 68.2 ± 2.8%, intermediate response (IR, n = 6), TRI = 41.1 ± 4.2% and low response (LR, n = 2), TRI = 13.4 ± 14.3%, producing therapy response categorization which had not been fully registered in single-slice studies. Results agreed with the multi-slice approach being feasible and producing an inverse correlation between TRI and Ki67 immunostaining. Additionally, ca. 7-day oscillations of TRI were observed, suggesting that host immune system activation in response to treatment could contribute to the responding patterns detected.

SELECTION OF CITATIONS
SEARCH DETAIL
...