12 research outputs found

    Patient-Specific Metrics of Invasiveness Reveal Significant Prognostic Benefit of Resection in a Predictable Subset of Gliomas

    Get PDF
    Object Malignant gliomas are incurable, primary brain neoplasms noted for their potential to extensively invade brain parenchyma. Current methods of clinical imaging do not elucidate the full extent of brain invasion, making it difficult to predict which, if any, patients are likely to benefit from gross total resection. Our goal was to apply a mathematical modeling approach to estimate the overall tumor invasiveness on a patient-by-patient basis and determine whether gross total resection would improve survival in patients with relatively less invasive gliomas. Methods In 243 patients presenting with contrast-enhancing gliomas, estimates of the relative invasiveness of each patient's tumor, in terms of the ratio of net proliferation rate of the glioma cells to their net dispersal rate, were derived by applying a patient-specific mathematical model to routine pretreatment MR imaging. The effect of varying degrees of extent of resection on overall survival was assessed for cohorts of patients grouped by tumor invasiveness. Results We demonstrate that patients with more diffuse tumors showed no survival benefit (Pā€Š=ā€Š0.532) from gross total resection over subtotal/biopsy, while those with nodular (less diffuse) tumors showed a significant benefit (Pā€Š=ā€Š0.00142) with a striking median survival benefit of over eight months compared to sub-totally resected tumors in the same cohort (an 80% improvement in survival time for GTR only seen for nodular tumors). Conclusions These results suggest that our patient-specific, model-based estimates of tumor invasiveness have clinical utility in surgical decision making. Quantification of relative invasiveness assessed from routinely obtained pre-operative imaging provides a practical predictor of the benefit of gross total resection

    Multiparameter MRI Predictors of Long-Term Survival in Glioblastoma Multiforme

    No full text
    Standard-of-care multiparameter magnetic resonance imaging (MRI) scans of the brain were used to objectively subdivide glioblastoma multiforme (GBM) tumors into regions that correspond to variations in blood flow, interstitial edema, and cellular density. We hypothesized that the distribution of these distinct tumor ecological ā€œhabitatsā€ at the time of presentation will impact the course of the disease. We retrospectively analyzed initial MRI scans in 2 groups of patients diagnosed with GBM, a long-term survival group comprising subjects who survived >36 month postdiagnosis, and a short-term survival group comprising subjects who survived ā‰¤19 month postdiagnosis. The single-institution discovery cohort contained 22 subjects in each group, while the multi-institution validation cohort contained 15 subjects per group. MRI voxel intensities were calibrated, and tumor voxels clustered on contrast-enhanced T1-weighted and fluid-attenuated inversion-recovery (FLAIR) images into 6 distinct ā€œhabitatsā€ based on low- to medium- to high-contrast enhancement and lowā€“high signal on FLAIR scans. Habitat 6 (high signal on calibrated contrast-enhanced T1-weighted and FLAIR sequences) comprised a significantly higher volume fraction of tumors in the long-term survival group (discovery cohort, 35% Ā± 6.5%; validation cohort, 34% Ā± 4.8%) compared with tumors in the short-term survival group (discovery cohort, 17% Ā± 4.5%, p < 0.03; validation cohort, 16 Ā± 4.0%, p < 0.007). Of the 6 distinct MRI-defined habitats, the fractional tumor volume of habitat 6 at diagnosis was significantly predictive of long- or short-term survival. We discuss a possible mechanistic basis for this association and implications for habitat-driven adaptive therapy of GBM

    Distinct Phenotypic Clusters of Glioblastoma Growth and Response Kinetics Predict Survival

    No full text
    PURPOSE: Despite the intra- and intertumoral heterogeneity seen in glioblastoma multiforme (GBM), there is little definitive data on the underlying cause of the differences in patient survivals. Serial imaging assessment of tumor growth allows quantification of tumor growth kinetics (TGK) measured in terms of changes in the velocity of radial expansion seen on imaging. Because a systematic study of this entire TGK phenotype-growth before treatment and during each treatment to recurrence -has never been coordinately studied in GBMs, we sought to identify whether patients cluster into discrete groups on the basis of their TGK. PATIENTS AND METHODS: From our multi-institutional database, we identified 48 patients who underwent maximally safe resection followed by radiotherapy with imaging follow-up through the time of recurrence. The patients were then clustered into two groups through a k-means algorithm taking as input only the TGK before and during treatment. RESULTS: There was a significant survival difference between the clusters ( P = .003). Paradoxically, patients among the long-lived cluster had significantly larger tumors at diagnosis ( P = .027) and faster growth before treatment ( P = .003) but demonstrated a better response to adjuvant chemotherapy ( P = .048). A predictive model was built to identify which cluster patients would likely fall into on the basis of information that would be available to clinicians immediately after radiotherapy (accuracy, 90.3%). CONCLUSION: Dichotomizing the heterogeneity of GBMs into two populations-one faster growing yet more responsive with increased survival and one slower growing yet less responsive with shorter survival-suggests that many patients who receive standard-of-care treatments may get better benefit from select alternative treatments

    Uncertainty quantification in the radiogenomics modeling of EGFR amplification in glioblastoma

    No full text
    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 tumorā€”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

    Ļ/D Assessment.

    No full text
    <p>This figure presents an overview of how the ā€œrelative invasiveness,ā€ or Ļ/D, is obtained. Tumor volumes are segmented from T1Gd and T2 MRI. The measured volume is approximated with a sphere in order to obtain a radius. The T1Gd and T2 radii are associated with different levels of detection, with T2 at low tumor cell density and T1Gd abnormality associated with high tumor cell density. The relationship between these two radii describes the steepness of the tumor cell profile, or ā€œrelative invasiveness.ā€</p

    Results of iterative Kaplan-Meier Analysis in each invasiveness cohort.

    No full text
    <p>Number of cells remaining was calculated for each patient, based on their Ļ/D and measured residual enhancing disease. Each possible threshold was iterated through to separate the patients into large and small residual tumor cell population cohorts. White boxes correspond to thresholds separating patients into groups with significantly different (p<0.05) survival. White stars indicate tests with no p-value, as the threshold did not separate the patients in the given invasiveness cohort into two groups. Black asterisks indicate tests with p<0.05. Black bins with white x's indicate no the threshold did not separate the patients in the given invasiveness cohort into two groups. For example, for the diffuse case (<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0099057#pone-0099057-g003" target="_blank">Figure 3</a>, top row), the black bins with white x's represent the fact that even GTR was unable to achieve a remaining cell burden less than the cutoff (up to approximately 10<sup>9</sup>). Further, amongst the most diffuse gliomas, no threshold for a residual cells following resection was found to be significant of outcome represented visually as the lack of a white bar in the top row. Although less dramatic, the moderate cohort was unable to equate a GTR with <10<sup>8.5</sup> cells remaining represented by the black bars with white x's to the left on middle row of <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0099057#pone-0099057-g003" target="_blank">Figure 3</a>. While resection of tumors in the nodular cohort were able to attain residual disease burdens at all levels down to <10<sup>7</sup> cells.</p

    Patient-Specific Simulations of Tumor Cell Distribution and Density for both a Relatively Diffuse and a Relatively Nodular Glioblastoma.

    No full text
    <p>T1Gd and T2 MRIs for two newly diagnosed glioblastoma patients, one relatively diffuse with a low Ļ/D (a,c) and one more nodular with a high Ļ/D (b,d). A simulation of the diffuse glioma extent predicted by the patient-specific simulation for the diffuse (low Ļ/D) patient (e) and the more nodular (high Ļ/D) patient (f) is overlayed on the T1Gd MRI with red and blue indicating high and low (but nonzero) glioma cell density, respectively. The effect of GTR is shown as a black region with a white outline and highlights the significant diffuse extent of glioma cells remaining post-GTR. In the more nodular (high Ļ/D) case, GTR removes 75% of the pre-treatment glioma cells leaving 8.4e8 cells while in the diffusely invasive (low Ļ/D) case, GTR removes only 27% of the pre-treatment glioma cells leaving 4.2e9 cells, an order of magnitude higher than the nodular case. The large number of tumor cells remaining after resection of a diffuse tumor drives recurrence.</p

    Clinical Data Table.

    No full text
    <p>Median age and range, distribution of males and females, race (unknown, Caucasian, Asian, Hispanic, Black), median KPS and range, median XRT Dose and range, number of patients diagnosed in the 90's, number of patients diagnosed in 2000 or later, number receiving preoperative steroids, and number of patients who received concurrent Temozolomide with XRT are shown. Proportion chi-square tests were performed to compare steroid administration, concurrent TMZ, and proportion of patients who received GTR vs. STR/Bx between the three invasiveness cohorts. No statistical difference at the Pā€Š=ā€Š0.05 significance level was found in any of these variables between cohorts. ANOVA tests were performed to compare KPS scores, age, and XRT doses between invasiveness cohorts. No difference at the Pā€Š=ā€Š0.05 significance level was found in any of these variables between cohorts.</p><p>* - Indicates no significant difference (pā‰¤0.05) between cohorts in this variable, per Proportion Chi-square test.</p><p>** - Indicates no significant difference (pā‰¤0.05) between cohorts in this variable, per ANOVA test.</p><p>Clinical Data Table.</p

    Image-localized biopsy mapping of brain tumor heterogeneity: A single-center study protocol.

    No full text
    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
    corecore