7
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Development and validation of an interpretable deep learning framework for Alzheimer’s disease classification

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Qiu et al. present a novel deep learning strategy to generate high-resolution visualizations of Alzheimer’s disease risk in humans that are highly interpretable and can accurately predict Alzheimer’s disease status. They then test the model by comparing its performance to that of neurologists and neuropathological data.

          Abstract

          Alzheimer’s disease is the primary cause of dementia worldwide, with an increasing morbidity burden that may outstrip diagnosis and management capacity as the population ages. Current methods integrate patient history, neuropsychological testing and MRI to identify likely cases, yet effective practices remain variably applied and lacking in sensitivity and specificity. Here we report an interpretable deep learning strategy that delineates unique Alzheimer’s disease signatures from multimodal inputs of MRI, age, gender, and Mini-Mental State Examination score. Our framework linked a fully convolutional network, which constructs high resolution maps of disease probability from local brain structure to a multilayer perceptron and generates precise, intuitive visualization of individual Alzheimer’s disease risk en route to accurate diagnosis. The model was trained using clinically diagnosed Alzheimer’s disease and cognitively normal subjects from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) dataset ( n = 417) and validated on three independent cohorts: the Australian Imaging, Biomarker and Lifestyle Flagship Study of Ageing (AIBL) ( n = 382), the Framingham Heart Study ( n = 102), and the National Alzheimer’s Coordinating Center (NACC) ( n = 582). Performance of the model that used the multimodal inputs was consistent across datasets, with mean area under curve values of 0.996, 0.974, 0.876 and 0.954 for the ADNI study, AIBL, Framingham Heart Study and NACC datasets, respectively. Moreover, our approach exceeded the diagnostic performance of a multi-institutional team of practicing neurologists ( n = 11), and high-risk cerebral regions predicted by the model closely tracked post-mortem histopathological findings. This framework provides a clinically adaptable strategy for using routinely available imaging techniques such as MRI to generate nuanced neuroimaging signatures for Alzheimer’s disease diagnosis, as well as a generalizable approach for linking deep learning to pathophysiological processes in human disease.

          Related collections

          Most cited references13

          • Record: found
          • Abstract: found
          • Article: not found

          Classification and mutation prediction from non–small cell lung cancer histopathology images using deep learning

          Visual inspection of histopathology slides is one of the main methods used by pathologists to assess the stage, type and subtype of lung tumors. Adenocarcinoma (LUAD) and squamous cell carcinoma (LUSC) are the most prevalent subtypes of lung cancer, and their distinction requires visual inspection by an experienced pathologist. In this study, we trained a deep convolutional neural network (inception v3) on whole-slide images obtained from The Cancer Genome Atlas to accurately and automatically classify them into LUAD, LUSC or normal lung tissue. The performance of our method is comparable to that of pathologists, with an average area under the curve (AUC) of 0.97. Our model was validated on independent datasets of frozen tissues, formalin-fixed paraffin-embedded tissues and biopsies. Furthermore, we trained the network to predict the ten most commonly mutated genes in LUAD. We found that six of them-STK11, EGFR, FAT1, SETBP1, KRAS and TP53-can be predicted from pathology images, with AUCs from 0.733 to 0.856 as measured on a held-out population. These findings suggest that deep-learning models can assist pathologists in the detection of cancer subtype or gene mutations. Our approach can be applied to any cancer type, and the code is available at https://github.com/ncoudray/DeepPATH .
            Bookmark
            • Record: found
            • Abstract: not found
            • Article: not found

            Visualizing data using ti-SNE

              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Neuroimaging correlates of pathologically defined subtypes of Alzheimer's disease: a case-control study.

              Three subtypes of Alzheimer's disease (AD) have been pathologically defined on the basis of the distribution of neurofibrillary tangles: typical AD, hippocampal-sparing AD, and limbic-predominant AD. Compared with typical AD, hippocampal-sparing AD has more neurofibrillary tangles in the cortex and fewer in the hippocampus, whereas the opposite pattern is seen in limbic-predominant AD. We aimed to determine whether MRI patterns of atrophy differ between these subtypes and whether structural neuroimaging could be a useful predictor of pathological subtype at autopsy. We identified patients who had been followed up in the Mayo Clinic Alzheimer's Disease Research Center (Rochester, MN, USA) or in the Alzheimer's Disease Patient Registry (Rochester, MN, USA) between 1992 and 2005. To be eligible for inclusion, participants had to have had dementia, AD pathology at autopsy (Braak stage ≥IV and intermediate to high probability of AD), and an ante-mortem MRI. Cases were assigned to one of three pathological subtypes--hippocampal-sparing, limbic-predominant, and typical AD--on the basis of neurofibrillary tangle counts in hippocampus and cortex and ratio of hippocampal to cortical burden, without reference to neuronal loss. Voxel-based morphometry and atlas-based parcellation were used to compare patterns of grey matter loss between groups and with age-matched control individuals. Neuroimaging was obtained at the time of first presentation. To summarise pair-wise group differences, we report the area under the receiver operator characteristic curve (AUROC). Of 177 eligible patients, 125 (71%) were classified as having typical AD, 33 (19%) as having limbic-predominant AD, and 19 (11%) as having hippocampal-sparing AD. Most patients with typical (98 [78%]) and limbic-predominant AD (31 [94%]) initially presented with an amnestic syndrome, but fewer patients with hippocampal-sparing AD (eight [42%]) did. The most severe medial temporal atrophy was recorded in patients with limbic-predominant AD, followed by those with typical disease, and then those with hippocampal-sparing AD. Conversely, the most severe cortical atrophy was noted in patients with hippocampal-sparing AD, followed by those with typical disease, and then limbic-predominant AD. The ratio of hippocampal to cortical volumes allowed the best discrimination between subtypes (p<0·0001; three-way AUROC 0·52 [95% CI 0·47-0·52]; ratio of AUROC to chance classification 3·1 [2·8-3·1]). Patients with typical AD and non-amnesic initial presentation had a significantly higher ratio of hippocampal to cortical volumes (median 0·045 [IQR 0·035-0·056]) than did those with an amnesic presentation (0·041 [0·031-0·057]; p=0·001). Patterns of atrophy on MRI differ across the pathological subtypes of AD. MRI regional volumetric analysis can reliably track the distribution of neurofibrillary tangle pathology and can predict pathological subtype of AD at autopsy. US National Institutes of Health (National Institute on Aging). Copyright © 2012 Elsevier Ltd. All rights reserved.
                Bookmark

                Author and article information

                Journal
                Brain
                Brain
                brainj
                Brain
                Oxford University Press
                0006-8950
                1460-2156
                June 2020
                01 May 2020
                01 May 2020
                : 143
                : 6
                : 1920-1933
                Affiliations
                [a1 ] Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine , Boston, MA, USA
                [a2 ] College of Arts and Sciences, Boston University , MA, USA
                [a3 ] Department of Anatomy and Neurobiology, Boston University School of Medicine , Boston, MA, USA
                [a4 ] The Framingham Heart Study, Boston University School of Medicine , Boston, MA, USA
                [a5 ] College of Computing, Georgia Institute of Technology , Atlanta, GA, USA
                [a6 ] Department of Neurology, Boston University School of Medicine , Boston, MA, USA
                [a7 ] Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences , Beijing, China
                [a8 ] Department of Neurology, University of Texas Health Science Center , Houston, TX, USA
                [a9 ] Department of Neurology, Texas Tech University Health Sciences Center , Lubbock, TX, USA
                [a10 ] Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center , Omaha, NE, USA
                [a11 ] Department of Epidemiology, Boston University School of Public Health , Boston, MA, USA
                [a12 ] Boston University Alzheimer’s Disease Center , Boston, MA, USA
                [a13 ] Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, MA, USA
                [a14 ] Hariri Institute for Computing and Computational Science & Engineering, Boston University , Boston, MA, USA
                Author notes
                Correspondence to: Vijaya B. Kolachalama, PhD 72 E. Concord Street, Evans 636, Boston, MA – 02118, USA E-mail: vkola@ 123456bu.edu

                Shangran Qiu, Prajakta S. Joshi, Matthew I. Miller and Chonghua Xue contributed equally to this work.

                Author information
                http://orcid.org/0000-0001-7367-5410
                http://orcid.org/0000-0002-7293-5556
                http://orcid.org/0000-0002-5312-8644
                Article
                awaa137
                10.1093/brain/awaa137
                7296847
                32357201
                8350d046-43af-4fde-b4ab-ba31b95a4e67
                © The Author(s) (2020). Published by Oxford University Press on behalf of the Guarantors of Brain.

                This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License ( http://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com

                History
                : 5 November 2019
                : 11 February 2020
                : 6 March 2020
                Page count
                Pages: 14
                Funding
                Funded by: National Center for Advancing Translational Sciences, DOI 10.13039/100006108;
                Funded by: National Institutes of Health, DOI 10.13039/100000002;
                Award ID: 1UL1TR001430
                Funded by: Scientist Development;
                Award ID: 17SDG33670323
                Funded by: American Heart Association, DOI 10.13039/100000968;
                Funded by: Hariri Research Award;
                Funded by: Hariri Institute for Computing and Computational Science & Engineering at Boston University;
                Funded by: Framingham Heart Study’s National Heart, Lung and Blood Institute;
                Award ID: N01-HC-25195
                Award ID: HHSN268201500001I
                Funded by: NIH, DOI 10.13039/100000002;
                Award ID: R56-AG062109
                Award ID: AG008122
                Award ID: R01-AG016495
                Award ID: R01-AG033040
                Funded by: Boston University’s Affinity Research Collaboratives;
                Funded by: Boston University Alzheimer’s Disease Center;
                Award ID: P30-AG013846
                Categories
                Original Articles

                Neurosciences
                dementia,biomarkers,alzheimer’s disease,structural mri,neurodegeneration
                Neurosciences
                dementia, biomarkers, alzheimer’s disease, structural mri, neurodegeneration

                Comments

                Comment on this article