62 research outputs found

    Generative discriminative models for multivariate inference and statistical mapping in medical imaging

    Full text link
    This paper presents a general framework for obtaining interpretable multivariate discriminative models that allow efficient statistical inference for neuroimage analysis. The framework, termed generative discriminative machine (GDM), augments discriminative models with a generative regularization term. We demonstrate that the proposed formulation can be optimized in closed form and in dual space, allowing efficient computation for high dimensional neuroimaging datasets. Furthermore, we provide an analytic estimation of the null distribution of the model parameters, which enables efficient statistical inference and p-value computation without the need for permutation testing. We compared the proposed method with both purely generative and discriminative learning methods in two large structural magnetic resonance imaging (sMRI) datasets of Alzheimer's disease (AD) (n=415) and Schizophrenia (n=853). Using the AD dataset, we demonstrated the ability of GDM to robustly handle confounding variations. Using Schizophrenia dataset, we demonstrated the ability of GDM to handle multi-site studies. Taken together, the results underline the potential of the proposed approach for neuroimaging analyses.Comment: To appear in MICCAI 2018 proceeding

    Robust automated detection of microstructural white matter degeneration in Alzheimer’s disease using machine learning classification of multicenter DTI data

    Get PDF
    Diffusion tensor imaging (DTI) based assessment of white matter fiber tract integrity can support the diagnosis of Alzheimer’s disease (AD). The use of DTI as a biomarker, however, depends on its applicability in a multicenter setting accounting for effects of different MRI scanners. We applied multivariate machine learning (ML) to a large multicenter sample from the recently created framework of the European DTI study on Dementia (EDSD). We hypothesized that ML approaches may amend effects of multicenter acquisition. We included a sample of 137 patients with clinically probable AD (MMSE 20.6±5.3) and 143 healthy elderly controls, scanned in nine different scanners. For diagnostic classification we used the DTI indices fractional anisotropy (FA) and mean diffusivity (MD) and, for comparison, gray matter and white matter density maps from anatomical MRI. Data were classified using a Support Vector Machine (SVM) and a Naïve Bayes (NB) classifier. We used two cross-validation approaches, (i) test and training samples randomly drawn from the entire data set (pooled cross-validation) and (ii) data from each scanner as test set, and the data from the remaining scanners as training set (scanner-specific cross-validation). In the pooled cross-validation, SVM achieved an accuracy of 80% for FA and 83% for MD. Accuracies for NB were significantly lower, ranging between 68% and 75%. Removing variance components arising from scanners using principal component analysis did not significantly change the classification results for both classifiers. For the scanner-specific cross-validation, the classification accuracy was reduced for both SVM and NB. After mean correction, classification accuracy reached a level comparable to the results obtained from the pooled cross-validation. Our findings support the notion that machine learning classification allows robust classification of DTI data sets arising from multiple scanners, even if a new data set comes from a scanner that was not part of the training sample

    Automatic ROI Selection in Structural Brain MRI Using SOM 3D Projection

    Get PDF
    This paper presents a method for selecting Regions of Interest (ROI) in brain Magnetic Resonance Imaging (MRI) for diagnostic purposes, using statistical learning and vector quantization techniques. The proposed method models the distribution of GM and WM tissues grouping the voxels belonging to each tissue in ROIs associated to a specific neurological disorder. Tissue distribution of normal and abnormal images is modelled by a Self-Organizing map (SOM), generating a set of representative prototypes, and the receptive field (RF) of each SOM prototype defines a ROI. Moreover, the proposed method computes the relative importance of each ROI by means of its discriminative power. The devised method has been assessed using 818 images from the Alzheimer's disease Neuroimaging Initiative (ADNI) which were previously segmented through Statistical Parametric Mapping (SPM). The proposed algorithm was used over these images to parcel ROIs associated to the Alzheimer's Disease (AD). Additionally, this method can be used to extract a reduced set of discriminative features for classification, since it compresses discriminative information contained in the brain. Voxels marked by ROIs which were computed using the proposed method, yield classification results up to 90% of accuracy for controls (CN) and Alzheimer's disease (AD) patients, and 84% of accuracy for Mild Cognitive Impairment (MCI) and AD patients.This work was partly supported by the MICINN under the TEC2012-34306 project and the Consejería de Innovación, Ciencia y Empresa (Junta de Andalucía, Spain) under the Excellence Projects P09-TIC-4530 and P11-TIC-7103. Data collection and sharing for this project was funded by the Alzheimer's Disease Neuroimaging Initiative (ADNI) (National Institutes of Health Grant U01 AG024904) and DOD ADNI (Department of Defense award number W81XWH-12-2-0012). ADNI is funded by the National Institute on Aging, the National Institute of Biomedical Imaging and Bioengineering, and through generous contributions from the following: Alzheimer's Association; Alzheimer's Drug Discovery Foundation; BioClinica, Inc.; Biogen Idec Inc.; Bristol-Myers Squibb Company; Eisai Inc.; Elan Pharmaceuticals, Inc.; Eli Lilly and Company; F. Hoffmann-La Roche Ltd and its affiliated company Genentech, Inc.; GE Healthcare; Innogenetics, N.V.; IXICO Ltd.; Janssen Alzheimer Immunotherapy Research & Development, LLC.; Johnson & Johnson Pharmaceutical Research & Development LLC.; Medpace, Inc.; Merck & Co., Inc.; Meso Scale Diagnostics, LLC.; NeuroRxResearch; Novartis Pharmaceuticals Corporation; Pfizer Inc.; Piramal Imaging; Servier; Synarc Inc.; and Takeda Pharmaceutical Company. The Canadian Institutes of Health Research is providing funds to support ADNI clinical sites in Canada. Private sector contributions are facilitated by the Foundation for the National Institutes of Health (www.fnih.org). The grantee organization is the Northern California Institute for Research and Education, and the study is coordinated by the Alzheimer's Disease Cooperative Study at the University of California, San Diego. ADNI data are disseminated by the Laboratory for Neuro Imaging at the University of Southern California

    A Wide and Deep Neural Network for Survival Analysis from Anatomical Shape and Tabular Clinical Data

    Full text link
    We introduce a wide and deep neural network for prediction of progression from patients with mild cognitive impairment to Alzheimer's disease. Information from anatomical shape and tabular clinical data (demographics, biomarkers) are fused in a single neural network. The network is invariant to shape transformations and avoids the need to identify point correspondences between shapes. To account for right censored time-to-event data, i.e., when it is only known that a patient did not develop Alzheimer's disease up to a particular time point, we employ a loss commonly used in survival analysis. Our network is trained end-to-end to combine information from a patient's hippocampus shape and clinical biomarkers. Our experiments on data from the Alzheimer's Disease Neuroimaging Initiative demonstrate that our proposed model is able to learn a shape descriptor that augments clinical biomarkers and outperforms a deep neural network on shape alone and a linear model on common clinical biomarkers.Comment: Data and Machine Learning Advances with Multiple Views Workshop, ECML-PKDD 201

    Multi-Method Analysis of MRI Images in Early Diagnostics of Alzheimer's Disease

    Get PDF
    The role of structural brain magnetic resonance imaging (MRI) is becoming more and more emphasized in the early diagnostics of Alzheimer's disease (AD). This study aimed to assess the improvement in classification accuracy that can be achieved by combining features from different structural MRI analysis techniques. Automatically estimated MR features used are hippocampal volume, tensor-based morphometry, cortical thickness and a novel technique based on manifold learning. Baseline MRIs acquired from all 834 subjects (231 healthy controls (HC), 238 stable mild cognitive impairment (S-MCI), 167 MCI to AD progressors (P-MCI), 198 AD) from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database were used for evaluation. We compared the classification accuracy achieved with linear discriminant analysis (LDA) and support vector machines (SVM). The best results achieved with individual features are 90% sensitivity and 84% specificity (HC/AD classification), 64%/66% (S-MCI/P-MCI) and 82%/76% (HC/P-MCI) with the LDA classifier. The combination of all features improved these results to 93% sensitivity and 85% specificity (HC/AD), 67%/69% (S-MCI/P-MCI) and 86%/82% (HC/P-MCI). Compared with previously published results in the ADNI database using individual MR-based features, the presented results show that a comprehensive analysis of MRI images combining multiple features improves classification accuracy and predictive power in detecting early AD. The most stable and reliable classification was achieved when combining all available features

    Learning Biomarker Models for Progression Estimation of Alzheimer’s Disease

    Get PDF
    Being able to estimate a patient’s progress in the course of Alzheimer’s disease and predicting future progression based on a number of observed biomarker values is of great interest for patients, clinicians and researchers alike. In this work, an approach for disease progress estimation is presented. Based on a set of subjects that convert to a more severe disease stage during the study, models that describe typical trajectories of biomarker values in the course of disease are learned using quantile regression. A novel probabilistic method is then derived to estimate the current disease progress as well as the rate of progression of an individual by fitting acquired biomarkers to the models. A particular strength of the method is its ability to naturally handle missing data. This means, it is applicable even if individual biomarker measurements are missing for a subject without requiring a retraining of the model. The functionality of the presented method is demonstrated using synthetic and—employing cognitive scores and image-based biomarkers—real data from the ADNI study. Further, three possible applications for progress estimation are demonstrated to underline the versatility of the approach: classification, construction of a spatio-temporal disease progression atlas and prediction of future disease progression

    Yet Another ADNI Machine Learning Paper? Paving The Way Towards Fully-reproducible Research on Classification of Alzheimer's Disease

    Get PDF
    International audienceIn recent years, the number of papers on Alzheimer's disease classification has increased dramatically, generating interesting methodological ideas on the use machine learning and feature extraction methods. However, practical impact is much more limited and, eventually, one could not tell which of these approaches are the most efficient. While over 90% of these works make use of ADNI an objective comparison between approaches is impossible due to variations in the subjects included, image pre-processing, performance metrics and cross-validation procedures. In this paper, we propose a framework for reproducible classification experiments using multimodal MRI and PET data from ADNI. The core components are: 1) code to automatically convert the full ADNI database into BIDS format; 2) a modular architecture based on Nipype in order to easily plug-in different classification and feature extraction tools; 3) feature extraction pipelines for MRI and PET data; 4) baseline classification approaches for unimodal and multimodal features. This provides a flexible framework for benchmarking different feature extraction and classification tools in a reproducible manner. Data management tools for obtaining the lists of subjects in AD, MCI converter, MCI non-converters, CN classes are also provided. We demonstrate its use on all (1519) baseline T1 MR images and all (1102) baseline FDG PET images from ADNI 1, GO and 2 with SPM-based feature extraction pipelines and three different classification techniques (linear SVM, anatomically regularized SVM and multiple kernel learning SVM). The highest accuracies achieved were: 91% for AD vs CN, 83% for MCIc vs CN, 75% for MCIc vs MCInc, 94% for AD-ABeta+ vs CN-ABeta- and 72% for MCIc-ABeta+ vs MCInc-ABeta+. The code will be made publicly available at the time of the conference

    Early prediction of Alzheimer's disease with non-local patch-based longitudinal descriptors

    No full text
    Comunicació presentada a: the 3rd International Workshop on Patch-based Techniques in Medical Imaging, amb conjunció amb MICCAI, celebrat a Québec, Canadà, el 14 de setembre de 2017.Alzheimer’s disease (AD) is characterized by a progressive decline in the cognitive functions accompanied by an atrophic process which can already be observed in the early stages using magnetic resonance images (MRI). Individualized prediction of future progression to AD, when patients are still in the mild cognitive impairment (MCI) stage, has potential impact for preventive treatment. Atrophy patterns extracted from longitudinal MRI sequences provide valuable information to identify MCI patients at higher risk of developing AD in the future. We present a novel descriptor that uses the similarity between local image patches to encode local displacements due to atrophy between a pair of longitudinal MRI scans. Using a conventional logistic regression classifier, our descriptor achieves 76% accuracy in predicting which MCI patients will progress to AD up to 3 years before conversion.The first author is co-financed by the Marie Curie FP7-PEOPLE-2012-COFUND 462 Action. Grant agreement no: 600387
    corecore