Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS One ; 19(4): e0299267, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38568950

RESUMEN

BACKGROUND AND OBJECTIVE: Glioblastoma (GBM) is one of the most aggressive and lethal human cancers. Intra-tumoral genetic heterogeneity poses a significant challenge for treatment. Biopsy is invasive, which motivates the development of non-invasive, MRI-based machine learning (ML) models to quantify intra-tumoral genetic heterogeneity for each patient. This capability holds great promise for enabling better therapeutic selection to improve patient outcome. METHODS: We proposed a novel Weakly Supervised Ordinal Support Vector Machine (WSO-SVM) to predict regional genetic alteration status within each GBM tumor using MRI. WSO-SVM was applied to a unique dataset of 318 image-localized biopsies with spatially matched multiparametric MRI from 74 GBM patients. The model was trained to predict the regional genetic alteration of three GBM driver genes (EGFR, PDGFRA and PTEN) based on features extracted from the corresponding region of five MRI contrast images. For comparison, a variety of existing ML algorithms were also applied. Classification accuracy of each gene were compared between the different algorithms. The SHapley Additive exPlanations (SHAP) method was further applied to compute contribution scores of different contrast images. Finally, the trained WSO-SVM was used to generate prediction maps within the tumoral area of each patient to help visualize the intra-tumoral genetic heterogeneity. RESULTS: WSO-SVM achieved 0.80 accuracy, 0.79 sensitivity, and 0.81 specificity for classifying EGFR; 0.71 accuracy, 0.70 sensitivity, and 0.72 specificity for classifying PDGFRA; 0.80 accuracy, 0.78 sensitivity, and 0.83 specificity for classifying PTEN; these results significantly outperformed the existing ML algorithms. Using SHAP, we found that the relative contributions of the five contrast images differ between genes, which are consistent with findings in the literature. The prediction maps revealed extensive intra-tumoral region-to-region heterogeneity within each individual tumor in terms of the alteration status of the three genes. CONCLUSIONS: This study demonstrated the feasibility of using MRI and WSO-SVM to enable non-invasive prediction of intra-tumoral regional genetic alteration for each GBM patient, which can inform future adaptive therapies for individualized oncology.


Asunto(s)
Glioblastoma , Humanos , Glioblastoma/diagnóstico por imagen , Glioblastoma/genética , Glioblastoma/patología , Medicina de Precisión , Heterogeneidad Genética , Imagen por Resonancia Magnética/métodos , Algoritmos , Aprendizaje Automático , Máquina de Vectores de Soporte , Receptores ErbB/genética
2.
Res Sq ; 2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38585856

RESUMEN

Intratumoral heterogeneity poses a significant challenge to the diagnosis and treatment of glioblastoma (GBM). This heterogeneity is further exacerbated during GBM recurrence, as treatment-induced reactive changes produce additional intratumoral heterogeneity that is ambiguous to differentiate on clinical imaging. There is an urgent need to develop non-invasive approaches to map the heterogeneous landscape of histopathological alterations throughout the entire lesion for each patient. We propose to predictively fuse Magnetic Resonance Imaging (MRI) with the underlying intratumoral heterogeneity in recurrent GBM using machine learning (ML) by leveraging image-localized biopsies with their associated locoregional MRI features. To this end, we develop BioNet, a biologically-informed neural network model, to predict regional distributions of three tissue-specific gene modules: proliferating tumor, reactive/inflammatory cells, and infiltrated brain tissue. BioNet offers valuable insights into the integration of multiple implicit and qualitative biological domain knowledge, which are challenging to describe in mathematical formulations. BioNet performs significantly better than a range of existing methods on cross-validation and blind test datasets. Voxel-level prediction maps of the gene modules by BioNet help reveal intratumoral heterogeneity, which can improve surgical targeting of confirmatory biopsies and evaluation of neuro-oncological treatment effectiveness. The non-invasive nature of the approach can potentially facilitate regular monitoring of the gene modules over time, and making timely therapeutic adjustment. These results also highlight the emerging role of ML in precision medicine.

3.
PLoS One ; 18(12): e0287767, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38117803

RESUMEN

Brain cancers pose a novel set of difficulties due to the limited accessibility of human brain tumor tissue. For this reason, clinical decision-making relies heavily on MR imaging interpretation, yet the mapping between MRI features and underlying biology remains ambiguous. Standard (clinical) tissue sampling fails to capture the full heterogeneity of the disease. Biopsies are required to obtain a pathological diagnosis and are predominantly taken from the tumor core, which often has different traits to the surrounding invasive tumor that typically leads to recurrent disease. One approach to solving this issue is to characterize the spatial heterogeneity of molecular, genetic, and cellular features of glioma through the intraoperative collection of multiple image-localized biopsy samples paired with multi-parametric MRIs. We have adopted this approach and are currently actively enrolling patients for our 'Image-Based Mapping of Brain Tumors' study. Patients are eligible for this research study (IRB #16-002424) if they are 18 years or older and undergoing surgical intervention for a brain lesion. Once identified, candidate patients receive dynamic susceptibility contrast (DSC) perfusion MRI and diffusion tensor imaging (DTI), in addition to standard sequences (T1, T1Gd, T2, T2-FLAIR) at their presurgical scan. During surgery, sample anatomical locations are tracked using neuronavigation. The collected specimens from this research study are used to capture the intra-tumoral heterogeneity across brain tumors including quantification of genetic aberrations through whole-exome and RNA sequencing as well as other tissue analysis techniques. To date, these data (made available through a public portal) have been used to generate, test, and validate predictive regional maps of the spatial distribution of tumor cell density and/or treatment-related key genetic marker status to identify biopsy and/or treatment targets based on insight from the entire tumor makeup. This type of methodology, when delivered within clinically feasible time frames, has the potential to further inform medical decision-making by improving surgical intervention, radiation, and targeted drug therapy for patients with glioma.


Asunto(s)
Neoplasias Encefálicas , Glioma , Humanos , Imagen de Difusión Tensora , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/genética , Glioma/diagnóstico por imagen , Glioma/genética , Glioma/patología , Imagen por Resonancia Magnética/métodos , Biopsia , Encéfalo/patología , Mapeo Encefálico
4.
Nat Commun ; 14(1): 6066, 2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37770427

RESUMEN

Sampling restrictions have hindered the comprehensive study of invasive non-enhancing (NE) high-grade glioma (HGG) cell populations driving tumor progression. Here, we present an integrated multi-omic analysis of spatially matched molecular and multi-parametric magnetic resonance imaging (MRI) profiling across 313 multi-regional tumor biopsies, including 111 from the NE, across 68 HGG patients. Whole exome and RNA sequencing uncover unique genomic alterations to unresectable invasive NE tumor, including subclonal events, which inform genomic models predictive of geographic evolution. Infiltrative NE tumor is alternatively enriched with tumor cells exhibiting neuronal or glycolytic/plurimetabolic cellular states, two principal transcriptomic pathway-based glioma subtypes, which respectively demonstrate abundant private mutations or enrichment in immune cell signatures. These NE phenotypes are non-invasively identified through normalized K2 imaging signatures, which discern cell size heterogeneity on dynamic susceptibility contrast (DSC)-MRI. NE tumor populations predicted to display increased cellular proliferation by mean diffusivity (MD) MRI metrics are uniquely associated with EGFR amplification and CDKN2A homozygous deletion. The biophysical mapping of infiltrative HGG potentially enables the clinical recognition of tumor subpopulations with aggressive molecular signatures driving tumor progression, thereby informing precision medicine targeting.


Asunto(s)
Productos Biológicos , Neoplasias Encefálicas , Glioma , Imágenes de Resonancia Magnética Multiparamétrica , Humanos , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Homocigoto , Eliminación de Secuencia , Glioma/diagnóstico por imagen , Glioma/genética , Glioma/patología , Imagen por Resonancia Magnética/métodos
5.
medRxiv ; 2023 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-37503239

RESUMEN

BACKGROUND: Glioblastoma is an extraordinarily heterogeneous tumor, yet the current treatment paradigm is a "one size fits all" approach. Hundreds of glioblastoma clinical trials have been deemed failures because they did not extend median survival, but these cohorts are comprised of patients with diverse tumors. Current methods of assessing treatment efficacy fail to fully account for this heterogeneity. METHODS: Using an image-based modeling approach, we predicted T-cell abundance from serial MRIs of patients enrolled in the dendritic cell (DC) vaccine clinical trial. T-cell predictions were quantified in both the contrast-enhancing and non-enhancing regions of the imageable tumor, and changes over time were assessed. RESULTS: A subset of patients in a DC vaccine clinical trial, who had previously gone undetected, were identified as treatment responsive and benefited from prolonged survival. A mere two months after initial vaccine administration, responsive patients had a decrease in model-predicted T-cells within the contrast-enhancing region, with a simultaneous increase in the T2/FLAIR region. CONCLUSIONS: In a field that has yet to see breakthrough therapies, these results highlight the value of machine learning in enhancing clinical trial assessment, improving our ability to prospectively prognosticate patient outcomes, and advancing the pursuit towards individualized medicine.

6.
medRxiv ; 2023 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-37333148

RESUMEN

Identification of key phenotypic regions such as necrosis, contrast enhancement, and edema on magnetic resonance imaging (MRI) is important for understanding disease evolution and treatment response in patients with glioma. Manual delineation is time intensive and not feasible for a clinical workflow. Automating phenotypic region segmentation overcomes many issues with manual segmentation, however, current glioma segmentation datasets focus on pre-treatment, diagnostic scans, where treatment effects and surgical cavities are not present. Thus, existing automatic segmentation models are not applicable to post-treatment imaging that is used for longitudinal evaluation of care. Here, we present a comparison of three-dimensional convolutional neural networks (nnU-Net architecture) trained on large temporally defined pre-treatment, post-treatment, and mixed cohorts. We used a total of 1563 imaging timepoints from 854 patients curated from 13 different institutions as well as diverse public data sets to understand the capabilities and limitations of automatic segmentation on glioma images with different phenotypic and treatment appearance. We assessed the performance of models using Dice coefficients on test cases from each group comparing predictions with manual segmentations generated by trained technicians. We demonstrate that training a combined model can be as effective as models trained on just one temporal group. The results highlight the importance of a diverse training set, that includes images from the course of disease and with effects from treatment, in the creation of a model that can accurately segment glioma MRIs at multiple treatment time points.

7.
IEEE Trans Autom Sci Eng ; 19(3): 2203-2215, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37700873

RESUMEN

The automated capability of generating spatial prediction for a variable of interest is desirable in various science and engineering domains. Take Precision Medicine of cancer as an example, in which the goal is to match patients with treatments based on molecular markers identified in each patient's tumor. A substantial challenge, however, is that the molecular markers can vary significantly at different spatial locations of a tumor. If this spatial distribution could be predicted, the precision of cancer treatment could be greatly improved by adapting treatment to the spatial molecular heterogeneity. This is a challenging task because no technology is available to measure the molecular markers at each spatial location within a tumor. Biopsy samples provide direct measurement, but they are scarce/local. Imaging, such as MRI, is global, but it only provides proxy/indirect measurement. Also available are mechanistic models or domain knowledge, which are often approximate or incomplete. This paper proposes a novel machine learning framework to fuse the three sources of data/information to generate spatial prediction, namely the knowledge-infused global-local data fusion (KGL) model. A novel mathematical formulation is proposed and solved with theoretical study. We present a real-data application of predicting the spatial distribution of Tumor Cell Density (TCD)-an important molecular marker for brain cancer. A total of 82 biopsy samples were acquired from 18 patients with glioblastoma, together with 6 MRI contrast images from each patient and biological knowledge encoded by a PDE simulator-based mechanistic model called Proliferation-Invasion (PI). KGL achieved the highest prediction accuracy and minimum prediction uncertainty compared with a variety of competing methods. The result has important implications for providing individualized, spatially-optimized treatment for each patient.

8.
Sci Rep ; 11(1): 3932, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33594116

RESUMEN

Radiogenomics uses machine-learning (ML) to directly connect the morphologic and physiological appearance of tumors on clinical imaging with underlying genomic features. Despite extensive growth in the area of radiogenomics across many cancers, and its potential role in advancing clinical decision making, no published studies have directly addressed uncertainty in these model predictions. We developed a radiogenomics ML model to quantify uncertainty using transductive Gaussian Processes (GP) and a unique dataset of 95 image-localized biopsies with spatially matched MRI from 25 untreated Glioblastoma (GBM) patients. The model generated predictions for regional EGFR amplification status (a common and important target in GBM) to resolve the intratumoral genetic heterogeneity across each individual tumor-a key factor for future personalized therapeutic paradigms. The model used probability distributions for each sample prediction to quantify uncertainty, and used transductive learning to reduce the overall uncertainty. We compared predictive accuracy and uncertainty of the transductive learning GP model against a standard GP model using leave-one-patient-out cross validation. Additionally, we used a separate dataset containing 24 image-localized biopsies from 7 high-grade glioma patients to validate the model. Predictive uncertainty informed the likelihood of achieving an accurate sample prediction. When stratifying predictions based on uncertainty, we observed substantially higher performance in the group cohort (75% accuracy, n = 95) and amongst sample predictions with the lowest uncertainty (83% accuracy, n = 72) compared to predictions with higher uncertainty (48% accuracy, n = 23), due largely to data interpolation (rather than extrapolation). On the separate validation set, our model achieved 78% accuracy amongst the sample predictions with lowest uncertainty. We present a novel approach to quantify radiogenomics uncertainty to enhance model performance and clinical interpretability. This should help integrate more reliable radiogenomics models for improved medical decision-making.


Asunto(s)
Genes erbB-1 , Glioblastoma/diagnóstico por imagen , Genómica de Imágenes , Aprendizaje Automático , Modelación Específica para el Paciente , Amplificación de Genes , Glioblastoma/genética , Humanos , Imagen por Resonancia Magnética , Incertidumbre
9.
Bull Math Biol ; 82(11): 143, 2020 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-33159592

RESUMEN

Glioblastoma (GBM) is the most aggressive primary brain tumor with a short median survival. Tumor recurrence is a clinical expectation of this disease and usually occurs along the resection cavity wall. However, previous clinical observations have suggested that in cases of ischemia following surgery, tumors are more likely to recur distally. Through the use of a previously established mechanistic model of GBM, the Proliferation Invasion Hypoxia Necrosis Angiogenesis (PIHNA) model, we explore the phenotypic drivers of this observed behavior. We have extended the PIHNA model to include a new nutrient-based vascular efficiency term that encodes the ability of local vasculature to provide nutrients to the simulated tumor. The extended model suggests sensitivity to a hypoxic microenvironment and the inherent migration and proliferation rates of the tumor cells are key factors that drive distal recurrence.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Isquemia , Modelos Biológicos , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Glioblastoma/patología , Humanos , Isquemia/complicaciones , Conceptos Matemáticos , Recurrencia Local de Neoplasia , Microambiente Tumoral
10.
Math Biosci Eng ; 17(5): 4905-4941, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-33120534

RESUMEN

Glioblastomas (GBMs) are the most aggressive primary brain tumours and have no known cure. Each individual tumour comprises multiple sub-populations of genetically-distinct cells that may respond differently to targeted therapies and may contribute to disappointing clinical trial results. Image-localized biopsy techniques allow multiple biopsies to be taken during surgery and provide information that identifies regions where particular sub-populations occur within an individual GBM, thus providing insight into their regional genetic variability. These sub-populations may also interact with one another in a competitive or cooperative manner; it is important to ascertain the nature of these interactions, as they may have implications for responses to targeted therapies. We combine genetic information from biopsies with a mechanistic model of interacting GBM sub-populations to characterise the nature of interactions between two commonly occurring GBM sub-populations, those with EGFR and PDGFRA genes amplified. We study population levels found across image-localized biopsy data from a cohort of 25 patients and compare this to model outputs under competitive, cooperative and neutral interaction assumptions. We explore other factors affecting the observed simulated sub-populations, such as selection advantages and phylogenetic ordering of mutations, which may also contribute to the levels of EGFR and PDGFRA amplified populations observed in biopsy data.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Neoplasias Encefálicas/genética , Glioblastoma/genética , Humanos , Mutación , Filogenia
11.
Bull Math Biol ; 82(9): 119, 2020 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-32909137

RESUMEN

Equation learning methods present a promising tool to aid scientists in the modeling process for biological data. Previous equation learning studies have demonstrated that these methods can infer models from rich datasets; however, the performance of these methods in the presence of common challenges from biological data has not been thoroughly explored. We present an equation learning methodology comprised of data denoising, equation learning, model selection and post-processing steps that infers a dynamical systems model from noisy spatiotemporal data. The performance of this methodology is thoroughly investigated in the face of several common challenges presented by biological data, namely, sparse data sampling, large noise levels, and heterogeneity between datasets. We find that this methodology can accurately infer the correct underlying equation and predict unobserved system dynamics from a small number of time samples when the data are sampled over a time interval exhibiting both linear and nonlinear dynamics. Our findings suggest that equation learning methods can be used for model discovery and selection in many areas of biology when an informative dataset is used. We focus on glioblastoma multiforme modeling as a case study in this work to highlight how these results are informative for data-driven modeling-based tumor invasion predictions.


Asunto(s)
Biología Computacional , Conceptos Matemáticos , Modelos Biológicos , Biología Computacional/métodos , Glioblastoma , Humanos , Aprendizaje , Dinámicas no Lineales
12.
BMC Cancer ; 20(1): 447, 2020 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-32429869

RESUMEN

BACKGROUND: Sex is recognized as a significant determinant of outcome among glioblastoma patients, but the relative prognostic importance of glioblastoma features has not been thoroughly explored for sex differences. METHODS: Combining multi-modal MR images, biomathematical models, and patient clinical information, this investigation assesses which pretreatment variables have a sex-specific impact on the survival of glioblastoma patients (299 males and 195 females). RESULTS: Among males, tumor (T1Gd) radius was a predictor of overall survival (HR = 1.027, p = 0.044). Among females, higher tumor cell net invasion rate was a significant detriment to overall survival (HR = 1.011, p < 0.001). Female extreme survivors had significantly smaller tumors (T1Gd) (p = 0.010 t-test), but tumor size was not correlated with female overall survival (p = 0.955 CPH). Both male and female extreme survivors had significantly lower tumor cell net proliferation rates than other patients (M p = 0.004, F p = 0.001, t-test). CONCLUSION: Despite similar distributions of the MR imaging parameters between males and females, there was a sex-specific difference in how these parameters related to outcomes.


Asunto(s)
Neoplasias Encefálicas/mortalidad , Glioblastoma/mortalidad , Imagen por Resonancia Magnética/métodos , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Niño , Femenino , Estudios de Seguimiento , Glioblastoma/patología , Glioblastoma/terapia , Humanos , Interpretación de Imagen Asistida por Computador , Masculino , Persona de Mediana Edad , Modelos Teóricos , Pronóstico , Estudios Retrospectivos , Factores Sexuales , Tasa de Supervivencia , Adulto Joven
13.
PLoS One ; 15(3): e0230492, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32218600

RESUMEN

BACKGROUND: Temozolomide (TMZ) has been the standard-of-care chemotherapy for glioblastoma (GBM) patients for more than a decade. Despite this long time in use, significant questions remain regarding how best to optimize TMZ therapy for individual patients. Understanding the relationship between TMZ response and factors such as number of adjuvant TMZ cycles, patient age, patient sex, and image-based tumor features, might help predict which GBM patients would benefit most from TMZ, particularly for those whose tumors lack O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation. METHODS AND FINDINGS: Using a cohort of 90 newly-diagnosed GBM patients treated according to the standard of care, we examined the relationships between several patient and tumor characteristics and volumetric and survival outcomes during adjuvant chemotherapy. Volumetric changes in MR imaging abnormalities during adjuvant therapy were used to assess TMZ response. T1Gd volumetric response is associated with younger patient age, increased number of TMZ cycles, longer time to nadir volume, and decreased tumor invasiveness. Moreover, increased adjuvant TMZ cycles corresponded with improved volumetric response only among more nodular tumors, and this volumetric response was associated with improved survival outcomes. Finally, in a subcohort of patients with known MGMT methylation status, methylated tumors were more diffusely invasive than unmethylated tumors, suggesting the improved response in nodular tumors is not driven by a preponderance of MGMT methylated tumors. CONCLUSIONS: Our finding that less diffusely invasive tumors are associated with greater volumetric response to TMZ suggests patients with these tumors may benefit from additional adjuvant TMZ cycles, even for those without MGMT methylation.


Asunto(s)
Neoplasias Encefálicas , Metilación de ADN/efectos de los fármacos , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/metabolismo , ADN de Neoplasias/metabolismo , Glioblastoma , Imagen por Resonancia Magnética , Regiones Promotoras Genéticas , Temozolomida/administración & dosificación , Proteínas Supresoras de Tumor/metabolismo , Adolescente , Adulto , Factores de Edad , Anciano , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Femenino , Glioblastoma/diagnóstico por imagen , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica
14.
Cancer Lett ; 477: 97-106, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32112907

RESUMEN

High-grade glioma (HGG), and particularly Glioblastoma (GBM), can exhibit pronounced intratumoral heterogeneity that confounds clinical diagnosis and management. While conventional contrast-enhanced MRI lacks the capability to resolve this heterogeneity, advanced MRI techniques and PET imaging offer a spectrum of physiologic and biophysical image features to improve the specificity of imaging diagnoses. Published studies have shown how integrating these advanced techniques can help better define histologically distinct targets for surgical and radiation treatment planning, and help evaluate the regional heterogeneity of tumor recurrence and response assessment following standard adjuvant therapy. Application of texture analysis and machine learning (ML) algorithms has also enabled the emerging field of radiogenomics, which can spatially resolve the regional and genetically distinct subpopulations that coexist within a single GBM tumor. This review focuses on the latest advances in neuro-oncologic imaging and their clinical applications for the assessment of intratumoral heterogeneity.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/patología , Glioma/diagnóstico por imagen , Glioma/patología , Algoritmos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Medios de Contraste , Glioma/genética , Glioma/terapia , Humanos , Aprendizaje Automático , Imagen por Resonancia Magnética/métodos , Recurrencia Local de Neoplasia/diagnóstico por imagen , Tomografía de Emisión de Positrones , Terapia Asistida por Computador
15.
Bull Math Biol ; 82(3): 43, 2020 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-32180054

RESUMEN

We analyze the wave speed of the Proliferation Invasion Hypoxia Necrosis Angiogenesis (PIHNA) model that was previously created and applied to simulate the growth and spread of glioblastoma (GBM), a particularly aggressive primary brain tumor. We extend the PIHNA model by allowing for different hypoxic and normoxic cell migration rates and study the impact of these differences on the wave-speed dynamics. Through this analysis, we find key variables that drive the outward growth of the simulated GBM. We find a minimum tumor wave-speed for the model; this depends on the migration and proliferation rates of the normoxic cells and is achieved under certain conditions on the migration rates of the normoxic and hypoxic cells. If the hypoxic cell migration rate is greater than the normoxic cell migration rate above a threshold, the wave speed increases above the predicted minimum. This increase in wave speed is explored through an eigenvalue and eigenvector analysis of the linearized PIHNA model, which yields an expression for this threshold. The PIHNA model suggests that an inherently faster-diffusing hypoxic cell population can drive the outward growth of a GBM as a whole, and that this effect is more prominent for faster-proliferating tumors that recover relatively slowly from a hypoxic phenotype. The findings presented here act as a first step in enabling patient-specific calibration of the PIHNA model.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Modelos Biológicos , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/fisiopatología , Recuento de Células , Movimiento Celular , Proliferación Celular , Simulación por Computador , Glioblastoma/irrigación sanguínea , Glioblastoma/fisiopatología , Humanos , Modelos Lineales , Conceptos Matemáticos , Necrosis , Invasividad Neoplásica , Neovascularización Patológica , Hipoxia Tumoral , Análisis de Ondículas
16.
PLoS Comput Biol ; 16(2): e1007672, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32101537

RESUMEN

Glioblastomas are aggressive primary brain tumors known for their inter- and intratumor heterogeneity. This disease is uniformly fatal, with intratumor heterogeneity the major reason for treatment failure and recurrence. Just like the nature vs nurture debate, heterogeneity can arise from intrinsic or environmental influences. Whilst it is impossible to clinically separate observed behavior of cells from their environmental context, using a mathematical framework combined with multiscale data gives us insight into the relative roles of variation from different sources. To better understand the implications of intratumor heterogeneity on therapeutic outcomes, we created a hybrid agent-based mathematical model that captures both the overall tumor kinetics and the individual cellular behavior. We track single cells as agents, cell density on a coarser scale, and growth factor diffusion and dynamics on a finer scale over time and space. Our model parameters were fit utilizing serial MRI imaging and cell tracking data from ex vivo tissue slices acquired from a growth-factor driven glioblastoma murine model. When fitting our model to serial imaging only, there was a spectrum of equally-good parameter fits corresponding to a wide range of phenotypic behaviors. When fitting our model using imaging and cell scale data, we determined that environmental heterogeneity alone is insufficient to match the single cell data, and intrinsic heterogeneity is required to fully capture the migration behavior. The wide spectrum of in silico tumors also had a wide variety of responses to an application of an anti-proliferative treatment. Recurrent tumors were generally less proliferative than pre-treatment tumors as measured via the model simulations and validated from human GBM patient histology. Further, we found that all tumors continued to grow with an anti-migratory treatment alone, but the anti-proliferative/anti-migratory combination generally showed improvement over an anti-proliferative treatment alone. Together our results emphasize the need to better understand the underlying phenotypes and tumor heterogeneity present in a tumor when designing therapeutic regimens.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/fisiopatología , Glioblastoma/diagnóstico por imagen , Glioblastoma/fisiopatología , Imagen por Resonancia Magnética , Animales , Proliferación Celular , Biología Computacional , Simulación por Computador , Humanos , Cinética , Masculino , Ratones Endogámicos NOD , Modelos Teóricos , Fenotipo , Ratas , Ratas Sprague-Dawley
17.
Sci Rep ; 9(1): 10063, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31296889

RESUMEN

Glioblastoma (GBM) is a heterogeneous and lethal brain cancer. These tumors are followed using magnetic resonance imaging (MRI), which is unable to precisely identify tumor cell invasion, impairing effective surgery and radiation planning. We present a novel hybrid model, based on multiparametric intensities, which combines machine learning (ML) with a mechanistic model of tumor growth to provide spatially resolved tumor cell density predictions. The ML component is an imaging data-driven graph-based semi-supervised learning model and we use the Proliferation-Invasion (PI) mechanistic tumor growth model. We thus refer to the hybrid model as the ML-PI model. The hybrid model was trained using 82 image-localized biopsies from 18 primary GBM patients with pre-operative MRI using a leave-one-patient-out cross validation framework. A Relief algorithm was developed to quantify relative contributions from the data sources. The ML-PI model statistically significantly outperformed (p < 0.001) both individual models, ML and PI, achieving a mean absolute predicted error (MAPE) of 0.106 ± 0.125 versus 0.199 ± 0.186 (ML) and 0.227 ± 0.215 (PI), respectively. Associated Pearson correlation coefficients for ML-PI, ML, and PI were 0.838, 0.518, and 0.437, respectively. The Relief algorithm showed the PI model had the greatest contribution to the result, emphasizing the importance of the hybrid model in achieving the high accuracy.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Glioblastoma/diagnóstico por imagen , Imágenes de Resonancia Magnética Multiparamétrica/métodos , Algoritmos , Recuento de Células , Humanos , Interpretación de Imagen Asistida por Computador , Aprendizaje Automático , Modelos Estadísticos , Modelos Teóricos , Pronóstico
18.
Am J Clin Oncol ; 42(8): 655-661, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31343422

RESUMEN

Although glioblastoma (GBM) is a fatal primary brain cancer with short median survival of 15 months, a small number of patients survive >5 years after diagnosis; they are known as extreme survivors (ES). Because of their rarity, very little is known about what differentiates these outliers from other patients with GBM. For the purpose of identifying unknown drivers of extreme survivorship in GBM, the ENDURES consortium (ENvironmental Dynamics Underlying Responsive Extreme Survivors of GBM) was developed. This consortium is a multicenter collaborative network of investigators focused on the integration of multiple types of clinical data and the creation of patient-specific models of tumor growth informed by radiographic and histologic parameters. Leveraging our combined resources, the goals of the ENDURES consortium are 2-fold: (1) to build a curated, searchable, multilayered repository housing clinical and outcome data on a large cohort of ES patients with GBM; and (2) to leverage the ENDURES repository for new insights into tumor behavior and novel targets for prolonging survival for all patients with GBM. In this article, the authors review the available literature and discuss what is already known about ES. The authors then describe the creation of their consortium and some preliminary results.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Sistema de Registros , Anciano , Neoplasias Encefálicas/patología , Femenino , Glioblastoma/patología , Humanos , Gestión de la Información , Comunicación Interdisciplinaria , Colaboración Intersectorial , Masculino , Persona de Mediana Edad , Sistema de Registros/normas , Tasa de Supervivencia
19.
Phys Biol ; 16(4): 041005, 2019 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-30991381

RESUMEN

Whether the nom de guerre is Mathematical Oncology, Computational or Systems Biology, Theoretical Biology, Evolutionary Oncology, Bioinformatics, or simply Basic Science, there is no denying that mathematics continues to play an increasingly prominent role in cancer research. Mathematical Oncology-defined here simply as the use of mathematics in cancer research-complements and overlaps with a number of other fields that rely on mathematics as a core methodology. As a result, Mathematical Oncology has a broad scope, ranging from theoretical studies to clinical trials designed with mathematical models. This Roadmap differentiates Mathematical Oncology from related fields and demonstrates specific areas of focus within this unique field of research. The dominant theme of this Roadmap is the personalization of medicine through mathematics, modelling, and simulation. This is achieved through the use of patient-specific clinical data to: develop individualized screening strategies to detect cancer earlier; make predictions of response to therapy; design adaptive, patient-specific treatment plans to overcome therapy resistance; and establish domain-specific standards to share model predictions and to make models and simulations reproducible. The cover art for this Roadmap was chosen as an apt metaphor for the beautiful, strange, and evolving relationship between mathematics and cancer.


Asunto(s)
Matemática/métodos , Oncología Médica/métodos , Biología de Sistemas/métodos , Biología Computacional , Simulación por Computador , Humanos , Modelos Biológicos , Modelos Teóricos , Neoplasias/diagnóstico , Neoplasias/terapia , Análisis de la Célula Individual/métodos
20.
Math Biosci ; 312: 59-66, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31009624

RESUMEN

Kinetic parameter estimates for mathematical models of glioblastoma multiforme (GBM), derived from clinical scans, have been used to predict the occurrence of hypoxia, necrosis, response to radiation therapy, and overall survival. Modeling GBM growth in a cerebral model encounters anatomical boundaries that interfere with model calibration from clinical measurements. METHODS: The effect of boundaries is examined on both spherically symmetric and anatomical models of tumor growth. This effect is incorporated into a method that updates kinetic parameters. The efficacy of this method in reproducing clinical image-derived subject data is evaluated. RESULTS: Spherically symmetric simulations of tumor growth with simple boundaries behave predictably when in a linear phase of growth. Anatomic simulations of eleven out of twenty subjects demonstrated improved fit to subject data with the new method. When only subjects exhibiting linear growth are considered, eight out of nine subject demonstrate improved fit to the data. CONCLUSION: Anatomical boundaries to tumor growth measurably deflect progression and affect estimates of kinetic parameters. The presented method reliably updates kinetic parameters to fit anatomic computational models to clinically derived subject data when those data are in a linear regime.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Modelos Biológicos , Neoplasias Encefálicas/diagnóstico , Glioblastoma/diagnóstico , Humanos , Pronóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA