Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Genome Med ; 16(1): 51, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38566128

ABSTRACT

BACKGROUND: A major contributing factor to glioblastoma (GBM) development and progression is its ability to evade the immune system by creating an immune-suppressive environment, where GBM-associated myeloid cells, including resident microglia and peripheral monocyte-derived macrophages, play critical pro-tumoral roles. However, it is unclear whether recruited myeloid cells are phenotypically and functionally identical in GBM patients and whether this heterogeneity is recapitulated in patient-derived orthotopic xenografts (PDOXs). A thorough understanding of the GBM ecosystem and its recapitulation in preclinical models is currently missing, leading to inaccurate results and failures of clinical trials. METHODS: Here, we report systematic characterization of the tumor microenvironment (TME) in GBM PDOXs and patient tumors at the single-cell and spatial levels. We applied single-cell RNA sequencing, spatial transcriptomics, multicolor flow cytometry, immunohistochemistry, and functional studies to examine the heterogeneous TME instructed by GBM cells. GBM PDOXs representing different tumor phenotypes were compared to glioma mouse GL261 syngeneic model and patient tumors. RESULTS: We show that GBM tumor cells reciprocally interact with host cells to create a GBM patient-specific TME in PDOXs. We detected the most prominent transcriptomic adaptations in myeloid cells, with brain-resident microglia representing the main population in the cellular tumor, while peripheral-derived myeloid cells infiltrated the brain at sites of blood-brain barrier disruption. More specifically, we show that GBM-educated microglia undergo transition to diverse phenotypic states across distinct GBM landscapes and tumor niches. GBM-educated microglia subsets display phagocytic and dendritic cell-like gene expression programs. Additionally, we found novel microglial states expressing cell cycle programs, astrocytic or endothelial markers. Lastly, we show that temozolomide treatment leads to transcriptomic plasticity and altered crosstalk between GBM tumor cells and adjacent TME components. CONCLUSIONS: Our data provide novel insights into the phenotypic adaptation of the heterogeneous TME instructed by GBM tumors. We show the key role of microglial phenotypic states in supporting GBM tumor growth and response to treatment. Our data place PDOXs as relevant models to assess the functionality of the TME and changes in the GBM ecosystem upon treatment.


Subject(s)
Brain Neoplasms , Glioblastoma , Mice , Animals , Humans , Glioblastoma/genetics , Glioblastoma/metabolism , Microglia/metabolism , Ecosystem , Heterografts , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Phenotype , Disease Models, Animal , Dendritic Cells/metabolism , Tumor Microenvironment/genetics
2.
Brain Sci ; 14(4)2024 Mar 23.
Article in English | MEDLINE | ID: mdl-38671953

ABSTRACT

Raman spectroscopy (RS) has demonstrated its utility in neurooncological diagnostics, spanning from intraoperative tumor detection to the analysis of tissue samples peri- and postoperatively. In this study, we employed Raman spectroscopy (RS) to monitor alterations in the molecular vibrational characteristics of a broad range of formalin-fixed, paraffin-embedded (FFPE) intracranial neoplasms (including primary brain tumors and meningiomas, as well as brain metastases) and considered specific challenges when employing RS on FFPE tissue during the routine neuropathological workflow. We spectroscopically measured 82 intracranial neoplasms on CaF2 slides (in total, 679 individual measurements) and set up a machine learning framework to classify spectral characteristics by splitting our data into training cohorts and external validation cohorts. The effectiveness of our machine learning algorithms was assessed by using common performance metrics such as AUROC and AUPR values. With our trained random forest algorithms, we distinguished among various types of gliomas and identified the primary origin in cases of brain metastases. Moreover, we spectroscopically diagnosed tumor types by using biopsy fragments of pure necrotic tissue, a task unattainable through conventional light microscopy. In order to address misclassifications and enhance the assessment of our models, we sought out significant Raman bands suitable for tumor identification. Through the validation phase, we affirmed a considerable complexity within the spectroscopic data, potentially arising not only from the biological tissue subjected to a rigorous chemical procedure but also from residual components of the fixation and paraffin-embedding process. The present study demonstrates not only the potential applications but also the constraints of RS as a diagnostic tool in neuropathology, considering the challenges associated with conducting vibrational spectroscopic analysis on formalin-fixed, paraffin-embedded (FFPE) tissue.

3.
Sensors (Basel) ; 24(7)2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38610406

ABSTRACT

Wearable sensors could be beneficial for the continuous quantification of upper limb motor symptoms in people with Parkinson's disease (PD). This work evaluates the use of two inertial measurement units combined with supervised machine learning models to classify and predict a subset of MDS-UPDRS III subitems in PD. We attached the two compact wearable sensors on the dorsal part of each hand of 33 people with PD and 12 controls. Each participant performed six clinical movement tasks in parallel with an assessment of the MDS-UPDRS III. Random forest (RF) models were trained on the sensor data and motor scores. An overall accuracy of 94% was achieved in classifying the movement tasks. When employed for classifying the motor scores, the averaged area under the receiver operating characteristic values ranged from 68% to 92%. Motor scores were additionally predicted using an RF regression model. In a comparative analysis, trained support vector machine models outperformed the RF models for specific tasks. Furthermore, our results surpass the literature in certain cases. The methods developed in this work serve as a base for future studies, where home-based assessments of pharmacological effects on motor function could complement regular clinical assessments.


Subject(s)
Parkinson Disease , Humans , Parkinson Disease/diagnosis , Machine Learning , Movement , Supervised Machine Learning , Hand
4.
Molecules ; 29(5)2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38474491

ABSTRACT

Understanding and classifying inherent tumor heterogeneity is a multimodal approach, which can be undertaken at the genetic, biochemical, or morphological level, among others. Optical spectral methods such as Raman spectroscopy aim at rapid and non-destructive tissue analysis, where each spectrum generated reflects the individual molecular composition of an examined spot within a (heterogenous) tissue sample. Using a combination of supervised and unsupervised machine learning methods as well as a solid database of Raman spectra of native glioblastoma samples, we succeed not only in distinguishing explicit tumor areas-vital tumor tissue and necrotic tumor tissue can correctly be predicted with an accuracy of 76%-but also in determining and classifying different spectral entities within the histomorphologically distinct class of vital tumor tissue. Measurements of non-pathological, autoptic brain tissue hereby serve as a healthy control since their respective spectroscopic properties form an individual and reproducible cluster within the spectral heterogeneity of a vital tumor sample. The demonstrated decipherment of a spectral glioblastoma heterogeneity will be valuable, especially in the field of spectroscopically guided surgery to delineate tumor margins and to assist resection control.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/pathology , Brain Neoplasms/pathology , Spectrum Analysis, Raman/methods , Machine Learning , Algorithms
5.
Molecules ; 29(5)2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38474679

ABSTRACT

Reliable training of Raman spectra-based tumor classifiers relies on a substantial sample pool. This study explores the impact of cryofixation (CF) and formalin fixation (FF) on Raman spectra using samples from surgery sites and a tumor bank. A robotic Raman spectrometer scans samples prior to the neuropathological analysis. CF samples showed no significant spectral deviations, appearance, or disappearance of peaks, but an intensity reduction during freezing and subsequent recovery during the thawing process. In contrast, FF induces sustained spectral alterations depending on molecular composition, albeit with good signal-to-noise ratio preservation. These observations are also reflected in the varying dual-class classifier performance, initially trained on native, unfixed samples: The Matthews correlation coefficient is 81.0% for CF and 58.6% for FF meningioma and dura mater. Training on spectral differences between original FF and pure formalin spectra substantially improves FF samples' classifier performance (74.2%). CF is suitable for training global multiclass classifiers due to its consistent spectrum shape despite intensity reduction. FF introduces changes in peak relationships while preserving the signal-to-noise ratio, making it more suitable for dual-class classification, such as distinguishing between healthy and malignant tissues. Pure formalin spectrum subtraction represents a possible method for mathematical elimination of the FF influence. These findings enable retrospective analysis of processed samples, enhancing pathological work and expanding machine learning techniques.


Subject(s)
Formaldehyde , Neoplasms , Humans , Retrospective Studies , Cryopreservation , Spectrum Analysis, Raman/methods
6.
Front Aging Neurosci ; 15: 1216163, 2023.
Article in English | MEDLINE | ID: mdl-37539346

ABSTRACT

Parkinson's disease (PD) is a progressive and complex neurodegenerative disorder associated with age that affects motor and cognitive functions. As there is currently no cure, early diagnosis and accurate prognosis are essential to increase the effectiveness of treatment and control its symptoms. Medical imaging, specifically magnetic resonance imaging (MRI), has emerged as a valuable tool for developing support systems to assist in diagnosis and prognosis. The current literature aims to improve understanding of the disease's structural and functional manifestations in the brain. By applying artificial intelligence to neuroimaging, such as deep learning (DL) and other machine learning (ML) techniques, previously unknown relationships and patterns can be revealed in this high-dimensional data. However, several issues must be addressed before these solutions can be safely integrated into clinical practice. This review provides a comprehensive overview of recent ML techniques analyzed for the automatic diagnosis and prognosis of PD in brain MRI. The main challenges in applying ML to medical diagnosis and its implications for PD are also addressed, including current limitations for safe translation into hospitals. These challenges are analyzed at three levels: disease-specific, task-specific, and technology-specific. Finally, potential future directions for each challenge and future perspectives are discussed.

7.
J Neurooncol ; 163(2): 327-338, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37237151

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is an aggressive brain cancer that typically results in death in the first 15 months after diagnosis. There have been limited advances in finding new treatments for GBM. In this study, we investigated molecular differences between patients with extremely short (≤ 9 months, Short term survivors, STS) and long survival (≥ 36 months, Long term survivors, LTS). METHODS: Patients were selected from an in-house cohort (GLIOTRAIN-cohort), using defined inclusion criteria (Karnofsky score > 70; age < 70 years old; Stupp protocol as first line treatment, IDH wild type), and a multi-omic analysis of LTS and STS GBM samples was performed. RESULTS: Transcriptomic analysis of tumour samples identified cilium gene signatures as enriched in LTS. Moreover, Immunohistochemical analysis confirmed the presence of cilia in the tumours of LTS. Notably, reverse phase protein array analysis (RPPA) demonstrated increased phosphorylated GAB1 (Y627), SRC (Y527), BCL2 (S70) and RAF (S338) protein expression in STS compared to LTS. Next, we identified 25 unique master regulators (MR) and 13 transcription factors (TFs) belonging to ontologies of integrin signalling and cell cycle to be upregulated in STS. CONCLUSION: Overall, comparison of STS and LTS GBM patients, identifies novel biomarkers and potential actionable therapeutic targets for the management of GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Aged , Glioblastoma/pathology , Prognosis , Brain Neoplasms/pathology , Brain/pathology , Survivors
8.
bioRxiv ; 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-36945572

ABSTRACT

Background: A major contributing factor to glioblastoma (GBM) development and progression is its ability to evade the immune system by creating an immune-suppressive environment, where GBM-associated myeloid cells, including resident microglia and peripheral monocyte-derived macrophages, play critical pro-tumoral roles. However, it is unclear whether recruited myeloid cells are phenotypically and functionally identical in GBM patients and whether this heterogeneity is recapitulated in patient-derived orthotopic xenografts (PDOXs). A thorough understanding of the GBM ecosystem and its recapitulation in preclinical models is currently missing, leading to inaccurate results and failures of clinical trials. Methods: Here, we report systematic characterization of the tumor microenvironment (TME) in GBM PDOXs and patient tumors at the single-cell and spatial levels. We applied single-cell RNA-sequencing, spatial transcriptomics, multicolor flow cytometry, immunohistochemistry and functional studies to examine the heterogeneous TME instructed by GBM cells. GBM PDOXs representing different tumor phenotypes were compared to glioma mouse GL261 syngeneic model and patient tumors. Results: We show that GBM tumor cells reciprocally interact with host cells to create a GBM patient-specific TME in PDOXs. We detected the most prominent transcriptomic adaptations in myeloid cells, with brain-resident microglia representing the main population in the cellular tumor, while peripheral-derived myeloid cells infiltrated the brain at sites of blood-brain barrier disruption. More specifically, we show that GBM-educated microglia undergo transition to diverse phenotypic states across distinct GBM landscapes and tumor niches. GBM-educated microglia subsets display phagocytic and dendritic cell-like gene expression programs. Additionally, we found novel microglial states expressing cell cycle programs, astrocytic or endothelial markers. Lastly, we show that temozolomide treatment leads to transcriptomic plasticity and altered crosstalk between GBM tumor cells and adjacent TME components. Conclusions: Our data provide novel insights into the phenotypic adaptation of the heterogeneous TME instructed by GBM tumors. We show the key role of microglial phenotypic states in supporting GBM tumor growth and response to treatment. Our data place PDOXs as relevant models to assess the functionality of the TME and changes in the GBM ecosystem upon treatment.

9.
Hum Brain Mapp ; 44(2): 762-778, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36250712

ABSTRACT

Segmenting deep brain structures from magnetic resonance images is important for patient diagnosis, surgical planning, and research. Most current state-of-the-art solutions follow a segmentation-by-registration approach, where subject magnetic resonance imaging (MRIs) are mapped to a template with well-defined segmentations. However, registration-based pipelines are time-consuming, thus, limiting their clinical use. This paper uses deep learning to provide a one-step, robust, and efficient deep brain segmentation solution directly in the native space. The method consists of a preprocessing step to conform all MRI images to the same orientation, followed by a convolutional neural network using the nnU-Net framework. We use a total of 14 datasets from both research and clinical collections. Of these, seven were used for training and validation and seven were retained for testing. We trained the network to segment 30 deep brain structures, as well as a brain mask, using labels generated from a registration-based approach. We evaluated the generalizability of the network by performing a leave-one-dataset-out cross-validation, and independent testing on unseen datasets. Furthermore, we assessed cross-domain transportability by evaluating the results separately on different domains. We achieved an average dice score similarity of 0.89 ± 0.04 on the test datasets when compared to the registration-based gold standard. On our test system, the computation time decreased from 43 min for a reference registration-based pipeline to 1.3 min. Our proposed method is fast, robust, and generalizes with high reliability. It can be extended to the segmentation of other brain structures. It is publicly available on GitHub, and as a pip package for convenient usage.


Subject(s)
Brain , Image Processing, Computer-Assisted , Humans , Image Processing, Computer-Assisted/methods , Reproducibility of Results , Brain/diagnostic imaging , Neural Networks, Computer , Magnetic Resonance Imaging/methods
10.
Sci Rep ; 12(1): 11465, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35794231

ABSTRACT

The study of complex diseases relies on large amounts of data to build models toward precision medicine. Such data acquisition is feasible in the context of high-throughput screening, in which the quality of the results relies on the accuracy of the image analysis. Although state-of-the-art solutions for image segmentation employ deep learning approaches, the high cost of manually generating ground truth labels for model training hampers the day-to-day application in experimental laboratories. Alternatively, traditional computer vision-based solutions do not need expensive labels for their implementation. Our work combines both approaches by training a deep learning network using weak training labels automatically generated with conventional computer vision methods. Our network surpasses the conventional segmentation quality by generalising beyond noisy labels, providing a 25% increase of mean intersection over union, and simultaneously reducing the development and inference times. Our solution was embedded into an easy-to-use graphical user interface that allows researchers to assess the predictions and correct potential inaccuracies with minimal human input. To demonstrate the feasibility of training a deep learning solution on a large dataset of noisy labels automatically generated by a conventional pipeline, we compared our solution against the common approach of training a model from a small manually curated dataset by several experts. Our work suggests that humans perform better in context interpretation, such as error assessment, while computers outperform in pixel-by-pixel fine segmentation. Such pipelines are illustrated with a case study on image segmentation for autophagy events. This work aims for better translation of new technologies to real-world settings in microscopy-image analysis.


Subject(s)
Deep Learning , High-Throughput Screening Assays , Autophagy , Humans , Image Processing, Computer-Assisted , Workflow
11.
Sci Rep ; 11(1): 23583, 2021 12 08.
Article in English | MEDLINE | ID: mdl-34880346

ABSTRACT

Meningiomas are among the most frequent tumors of the central nervous system. For a total resection, shown to decrease recurrences, it is paramount to reliably discriminate tumor tissue from normal dura mater intraoperatively. Raman spectroscopy (RS) is a non-destructive, label-free method for vibrational analysis of biochemical molecules. On the microscopic level, RS was already used to differentiate meningioma from dura mater. In this study we test its suitability for intraoperative macroscopic meningioma diagnostics. RS is applied to surgical specimen of intracranial meningiomas. The main purpose is the differentiation of tumor from normal dura mater, in order to potentially accelerate the diagnostic workflow. The collected meningioma and dura mater samples (n = 223 tissue samples from a total of 59 patients) are analyzed under untreated conditions using a new partially robotized RS acquisition system. Spectra (n = 1273) are combined with the according histopathological analysis for each sample. Based on this, a classifier is trained via machine learning. Our trained classifier separates meningioma and dura mater with a sensitivity of 96.06 [Formula: see text] 0.03% and a specificity of 95.44 [Formula: see text] 0.02% for internal fivefold cross validation and 100% and 93.97% if validated with an external test set. RS is an efficient method to discriminate meningioma from healthy dura mater in fresh tissue samples without additional processing or histopathological imaging. It is a quick and reliable complementary diagnostic tool to the neuropathological workflow and has potential for guided surgery. RS offers a safe way to examine unfixed surgical specimens in a perioperative setting.


Subject(s)
Dura Mater/pathology , Intraoperative Care/methods , Meningeal Neoplasms/diagnosis , Meningeal Neoplasms/pathology , Meningioma/diagnosis , Meningioma/pathology , Spectrum Analysis, Raman/methods , Cell Differentiation/physiology , Humans , Sensitivity and Specificity
12.
Biomed Tech (Berl) ; 66(6): 603-611, 2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34727584

ABSTRACT

Programming in deep brain stimulation (DBS) is a labour-intensive process for treating advanced motor symptoms. Specifically for patients with medication-refractory tremor in multiple sclerosis (MS). Wearable sensors are able to detect some manifestations of pathological signs, such as intention tremor in MS. However, methods are needed to visualise the response of tremor to DBS parameter changes in a clinical setting while patients perform the motor task finger-to-nose. To this end, we attended DBS programming sessions of a MS patient and intention tremor was effectively quantified by acceleration amplitude and frequency. A new method is introduced which results in the generation of therapeutic maps for a systematic review of the programming procedure in DBS. The maps visualise the combination of tremor acceleration power, clinical rating scores, total electrical energy delivered to the brain and possible side effects. Therapeutic maps have not yet been employed and could lead to a certain degree of standardisation for more objective decisions about DBS settings. The maps provide a base for future research on visualisation tools to assist physicians who frequently encounter patients for DBS therapy.


Subject(s)
Deep Brain Stimulation , Brain , Humans , Tremor/therapy
13.
Neurooncol Adv ; 3(1): vdab077, 2021.
Article in English | MEDLINE | ID: mdl-34355170

ABSTRACT

BACKGROUND: Although microscopic assessment is still the diagnostic gold standard in pathology, non-light microscopic methods such as new imaging methods and molecular pathology have considerably contributed to more precise diagnostics. As an upcoming method, Raman spectroscopy (RS) offers a "molecular fingerprint" that could be used to differentiate tissue heterogeneity or diagnostic entities. RS has been successfully applied on fresh and frozen tissue, however more aggressively, chemically treated tissue such as formalin-fixed, paraffin-embedded (FFPE) samples are challenging for RS. METHODS: To address this issue, we examined FFPE samples of morphologically highly heterogeneous glioblastoma (GBM) using RS in order to classify histologically defined GBM areas according to RS spectral properties. We have set up an SVM (support vector machine)-based classifier in a training cohort and corroborated our findings in a validation cohort. RESULTS: Our trained classifier identified distinct histological areas such as tumor core and necroses in GBM with an overall accuracy of 70.5% based on the spectral properties of RS. With an absolute misclassification of 21 out of 471 Raman measurements, our classifier has the property of precisely distinguishing between normal-appearing brain tissue and necrosis. When verifying the suitability of our classifier system in a second independent dataset, very little overlap between necrosis and normal-appearing brain tissue can be detected. CONCLUSION: These findings show that histologically highly variable samples such as GBM can be reliably recognized by their spectral properties using RS. As conclusion, we propose that RS may serve useful as a future method in the pathological toolbox.

14.
Free Neuropathol ; 22021 Jan.
Article in English | MEDLINE | ID: mdl-37284619

ABSTRACT

Objective and Methods: Timely discrimination between primary CNS lymphoma (PCNSL) and glioblastoma is crucial for diagnosis and therapy, but also determines the intraoperative surgical course. Advanced radiological methods allow for their distinction to a certain extent but ultimately, biopsies are still necessary for final diagnosis. As an upcoming method that enables tissue analysis by tracking changes in the vibrational state of molecules via inelastic scattered photons, we used Raman Spectroscopy (RS) as a label free method to examine specimens of both tumor entities intraoperatively, as well as postoperatively in formalin fixed paraffin embedded (FFPE) samples. Results: We applied and compared statistical performance of linear and nonlinear machine learning algorithms (Logistic Regression, Random Forest and XGBoost), and found that Random Forest classification distinguished the two tumor entities with a balanced accuracy of 82.4% in intraoperative tissue condition and with 94% using measurements of distinct tumor areas on FFPE tissue. Taking a deeper insight into the spectral properties of the tumor entities, we describe different tumor-specific Raman shifts of interest for classification. Conclusions: Due to our findings, we propose RS as an additional tool for fast and non-destructive tumor tissue discrimination, which may help to choose the proper treatment option. RS may further serve as a useful additional tool for neuropathological diagnostics with little requirements for tissue integrity.

15.
BMJ Case Rep ; 13(11)2020 Nov 30.
Article in English | MEDLINE | ID: mdl-33257397

ABSTRACT

Anorexia nervosa (AN) severely impacts individual's mental and physical health as well as quality of life. In 21% of cases no durable response to conservative treatment can be obtained. The serious course of the disease in the most severely affected patients justifies invasive treatment options. One of the treatment methods increasingly used in recent years is deep brain stimulation (DBS). A 42-year-old woman suffering from chronic AN of the bulimic subtype shows a 46.9% weight gain and a subjective increase in quality of life, 12 months after bilateral nucleus accumbens (NAcc) DBS implantation. No improvement in comorbid depression could be achieved. DBS of the NAcc is a treatment option to be considered in severe AN when conventional treatment modalities recommended by evidence-based guidelines have not been able to bring lasting relief to the patient's suffering.


Subject(s)
Anorexia Nervosa/therapy , Bulimia Nervosa/therapy , Deep Brain Stimulation , Weight Gain , Adult , Anorexia Nervosa/physiopathology , Anorexia Nervosa/psychology , Bulimia Nervosa/psychology , Electrodes, Implanted/adverse effects , Female , Humans , Quality of Life
16.
Acta Neuropathol ; 140(6): 919-949, 2020 12.
Article in English | MEDLINE | ID: mdl-33009951

ABSTRACT

Patient-based cancer models are essential tools for studying tumor biology and for the assessment of drug responses in a translational context. We report the establishment a large cohort of unique organoids and patient-derived orthotopic xenografts (PDOX) of various glioma subtypes, including gliomas with mutations in IDH1, and paired longitudinal PDOX from primary and recurrent tumors of the same patient. We show that glioma PDOXs enable long-term propagation of patient tumors and represent clinically relevant patient avatars that retain histopathological, genetic, epigenetic, and transcriptomic features of parental tumors. We find no evidence of mouse-specific clonal evolution in glioma PDOXs. Our cohort captures individual molecular genotypes for precision medicine including mutations in IDH1, ATRX, TP53, MDM2/4, amplification of EGFR, PDGFRA, MET, CDK4/6, MDM2/4, and deletion of CDKN2A/B, PTCH, and PTEN. Matched longitudinal PDOX recapitulate the limited genetic evolution of gliomas observed in patients following treatment. At the histological level, we observe increased vascularization in the rat host as compared to mice. PDOX-derived standardized glioma organoids are amenable to high-throughput drug screens that can be validated in mice. We show clinically relevant responses to temozolomide (TMZ) and to targeted treatments, such as EGFR and CDK4/6 inhibitors in (epi)genetically defined subgroups, according to MGMT promoter and EGFR/CDK status, respectively. Dianhydrogalactitol (VAL-083), a promising bifunctional alkylating agent in the current clinical trial, displayed high therapeutic efficacy, and was able to overcome TMZ resistance in glioblastoma. Our work underscores the clinical relevance of glioma organoids and PDOX models for translational research and personalized treatment studies and represents a unique publicly available resource for precision oncology.


Subject(s)
Brain Neoplasms/drug therapy , Glioma/drug therapy , Heterografts/immunology , Organoids/pathology , Temozolomide/therapeutic use , Animals , Brain Neoplasms/genetics , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioma/genetics , Heterografts/drug effects , Humans , Mice , Neoplasm Recurrence, Local/genetics , Organoids/immunology , Precision Medicine/methods , Rats
17.
Neuroimage ; 223: 117330, 2020 12.
Article in English | MEDLINE | ID: mdl-32890746

ABSTRACT

Deep brain stimulation (DBS) is a surgical therapy to alleviate symptoms of certain brain disorders by electrically modulating neural tissues. Computational models predicting electric fields and volumes of tissue activated are key for efficient parameter tuning and network analysis. Currently, we lack efficient and flexible software implementations supporting complex electrode geometries and stimulation settings. Available tools are either too slow (e.g. finite element method-FEM), or too simple, with limited applicability to basic use-cases. This paper introduces FastField, an efficient open-source toolbox for DBS electric field and VTA approximations. It computes scalable electric field approximations based on the principle of superposition, and VTA activation models from pulse width and axon diameter. In benchmarks and case studies, FastField is solved in about 0.2 s,  ~ 1000 times faster than using FEM. Moreover, it is almost as accurate as using FEM: average Dice overlap of 92%, which is around typical noise levels found in clinical data. Hence, FastField has the potential to foster efficient optimization studies and to support clinical applications.


Subject(s)
Brain/physiology , Deep Brain Stimulation , Electromagnetic Phenomena , Axons/physiology , Deep Brain Stimulation/instrumentation , Electrodes, Implanted , Electrophysiological Phenomena , Humans , Models, Neurological , Software
19.
Nat Commun ; 10(1): 1787, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30992437

ABSTRACT

The identity and unique capacity of cancer stem cells (CSC) to drive tumor growth and resistance have been challenged in brain tumors. Here we report that cells expressing CSC-associated cell membrane markers in Glioblastoma (GBM) do not represent a clonal entity defined by distinct functional properties and transcriptomic profiles, but rather a plastic state that most cancer cells can adopt. We show that phenotypic heterogeneity arises from non-hierarchical, reversible state transitions, instructed by the microenvironment and is predictable by mathematical modeling. Although functional stem cell properties were similar in vitro, accelerated reconstitution of heterogeneity provides a growth advantage in vivo, suggesting that tumorigenic potential is linked to intrinsic plasticity rather than CSC multipotency. The capacity of any given cancer cell to reconstitute tumor heterogeneity cautions against therapies targeting CSC-associated membrane epitopes. Instead inherent cancer cell plasticity emerges as a novel relevant target for treatment.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/genetics , Cell Plasticity/drug effects , Glioblastoma/genetics , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Biopsy , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Plasticity/genetics , Cohort Studies , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Temozolomide/pharmacology , Temozolomide/therapeutic use , Treatment Outcome , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
20.
Brain Stimul ; 11(4): 863-866, 2018.
Article in English | MEDLINE | ID: mdl-29429953

ABSTRACT

BACKGROUND: The gold standard for post-operative deep brain stimulation (DBS) parameter tuning is a monopolar review of all stimulation contacts, a strategy being challenged by recent developments of more complex electrode leads. OBJECTIVE: Providing a method to guide clinicians on DBS assessment and parameter tuning by automatically integrating patient individual data. METHODS: We present a fully automatic method for visualization of individual deep brain structures in relation to a DBS lead by combining precise electrode recovery from post-operative imaging with individual estimates of deep brain morphology utilizing a 7T-MRI deep brain atlas. RESULTS: The method was evaluated on 20 STN DBS cases. It demonstrated robust automatic creation of 3D-enabled PDF reports visualizing electrode to brain structure relations and proved valuable in detecting miss placed electrodes. DISCUSSION: Automatic DBS assessment is feasible and can conveniently provide clinicians with relevant information on DBS contact positions in relation to important anatomical structures.


Subject(s)
Deep Brain Stimulation/methods , Electronic Data Processing/methods , Postoperative Care/methods , Subthalamic Nucleus/physiology , Brain/physiology , Deep Brain Stimulation/instrumentation , Electrodes, Implanted , Humans , Magnetic Resonance Imaging/methods , Parkinson Disease/physiopathology , Parkinson Disease/therapy , Postoperative Care/instrumentation
SELECTION OF CITATIONS
SEARCH DETAIL
...