Analysing heterogeneity in Alzheimer Disease using multimodal normative modelling on ATN biomarkers

2404.05748

YC

0

Reddit

0

Published 4/10/2024 by Sayantan Kumara, Thomas Earnest, Braden Yang, Deydeep Kothapalli, Tammie L. S. Benzinger, Brian A. Gordon, Philip Payne, Aristeidis Sotiras

🧠

Abstract

Alzheimer Disease (AD) is a multi-faceted disorder, with each modality providing unique and complementary info about AD. In this study, we used a deep-learning based multimodal normative model to assess the heterogeneity in regional brain patterns for ATN (amyloid-tau-neurodegeneration) biomarkers. We selected discovery (n = 665) and replication (n = 430) cohorts with simultaneous availability of ATN biomarkers: Florbetapir amyloid, Flortaucipir tau and T1-weighted MRI (magnetic resonance imaging) imaging. A multimodal variational autoencoder (conditioned on age and sex) was used as a normative model to learn the multimodal regional brain patterns of a cognitively unimpaired (CU) control group. The trained model was applied on individuals on the ADS (AD Spectrum) to estimate their deviations (Z-scores) from the normative distribution, resulting in a Z-score regional deviation map per ADS individual per modality. ADS individuals with moderate or severe dementia showed higher proportion of regional outliers for each modality as well as more dissimilarity in modality-specific regional outlier patterns compared to ADS individuals with early or mild dementia. DSI was associated with the progressive stages of dementia, (ii) showed significant associations with neuropsychological composite scores and (iii) related to the longitudinal risk of CDR progression. Findings were reproducible in both discovery and replication cohorts. Our is the first study to examine the heterogeneity in AD through the lens of multiple neuroimaging modalities (ATN), based on distinct or overlapping patterns of regional outlier deviations. Regional MRI and tau outliers were more heterogenous than regional amyloid outliers. DSI has the potential to be an individual patient metric of neurodegeneration that can help in clinical decision making and monitoring patient response for anti-amyloid treatments.

Create account to get full access

or

If you already have an account, we'll log you in

Overview

  • This study used a deep learning-based multimodal normative model to assess the heterogeneity in regional brain patterns for Alzheimer's disease (AD) biomarkers.
  • The researchers selected cohorts with simultaneous availability of AD biomarkers: amyloid, tau, and structural MRI.
  • They used a multimodal variational autoencoder to learn the normative multimodal regional brain patterns in cognitively unimpaired individuals.
  • The trained model was then applied to individuals on the AD spectrum to estimate their deviations from the normative distribution, resulting in regional deviation maps.

Plain English Explanation

Alzheimer's disease is a complex disorder that affects the brain in various ways. In this study, the researchers used a deep learning approach to create a multimodal normative model that captures the typical patterns of amyloid, tau, and brain structure in healthy individuals. They then applied this model to people at different stages of Alzheimer's disease to see how their brain patterns deviated from the norm.

The key idea is that Alzheimer's affects different brain regions in different ways, and studying these regional differences can provide insights into the disease process. By using multiple brain imaging modalities (amyloid, tau, and MRI), the researchers were able to get a more comprehensive understanding of the brain changes associated with Alzheimer's.

The researchers found that individuals with more advanced dementia showed greater deviations from the normative brain patterns across all three modalities, and the regional outlier patterns were more distinct compared to those with early or mild dementia. This highlights the heterogeneous nature of Alzheimer's disease and suggests that studying multimodal brain patterns can provide valuable insights into the progression of the disease.

Technical Explanation

The researchers used a multimodal variational autoencoder as a normative model to learn the typical regional brain patterns of amyloid, tau, and structural MRI in a cohort of cognitively unimpaired individuals. They then applied this trained model to individuals across the Alzheimer's disease spectrum to estimate their deviations from the normative distribution, generating regional deviation maps for each modality.

The findings showed that individuals with moderate or severe dementia had a higher proportion of regional outliers across all three modalities, as well as more dissimilarity in the regional outlier patterns compared to those with early or mild dementia. These deviations from the normative multimodal brain patterns, referred to as the "Deviation Score Index" (DSI), were associated with the progressive stages of dementia, neuropsychological test scores, and the longitudinal risk of clinical decline.

This work is the first to examine the heterogeneity of Alzheimer's disease through the lens of multiple neuroimaging modalities, revealing distinct or overlapping patterns of regional outliers in amyloid, tau, and structural MRI. The researchers suggest that the DSI has the potential to be a useful individual-level metric of neurodegeneration that could aid in clinical decision-making and monitoring patient response to anti-amyloid treatments.

Critical Analysis

The researchers acknowledge several limitations in their study. First, the cohorts used for the discovery and replication analyses were relatively small, and the findings need to be validated in larger, more diverse populations. Additionally, the study was cross-sectional, and longitudinal data would be needed to fully understand the dynamic changes in multimodal brain patterns over the course of Alzheimer's disease.

Another potential limitation is the use of a single normative model, which may not capture the full range of normal variation in brain structure and function. Incorporating multiple normative models or using more advanced deep representation learning techniques could potentially improve the sensitivity and specificity of the deviation analyses.

It's also worth noting that the study focused on the regional patterns of amyloid, tau, and structural MRI, but other neuroimaging modalities, such as functional PET/MRI, could provide additional insights into the complex neural mechanisms underlying Alzheimer's disease.

Conclusion

This study demonstrates the potential of using a deep learning-based multimodal normative model to assess the heterogeneity of Alzheimer's disease across multiple neuroimaging modalities. The findings suggest that the Deviation Score Index, which captures the regional deviations from the normative brain patterns, could serve as a useful individual-level metric for monitoring disease progression and evaluating the effectiveness of therapeutic interventions. While further research is needed to validate and refine this approach, this work represents an important step towards a more comprehensive understanding of the complex neural underpinnings of Alzheimer's disease.



This summary was produced with help from an AI and may contain inaccuracies - check out the links to read the original source documents!

Related Papers

An interpretable generative multimodal neuroimaging-genomics framework for decoding Alzheimer's disease

An interpretable generative multimodal neuroimaging-genomics framework for decoding Alzheimer's disease

Giorgio Dolci (Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy, Tri-Institutional Center for Translational Research in Neuroimaging and Data Science), Federica Cruciani (Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy), Md Abdur Rahaman (Tri-Institutional Center for Translational Research in Neuroimaging and Data Science), Anees Abrol (Tri-Institutional Center for Translational Research in Neuroimaging and Data Science), Jiayu Chen (Tri-Institutional Center for Translational Research in Neuroimaging and Data Science), Zening Fu (Tri-Institutional Center for Translational Research in Neuroimaging and Data Science), Ilaria Boscolo Galazzo (Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy), Gloria Menegaz (Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy), Vince D. Calhoun (Tri-Institutional Center for Translational Research in Neuroimaging and Data Science)

YC

0

Reddit

0

Alzheimer's disease (AD) is the most prevalent form of dementia with a progressive decline in cognitive abilities. The AD continuum encompasses a prodormal stage known as Mild Cognitive Impairment (MCI), where patients may either progress to AD or remain stable. In this study, we leveraged structural and functional MRI to investigate the disease-induced grey matter and functional network connectivity changes. Moreover, considering AD's strong genetic component, we introduce SNPs as a third channel. Given such diverse inputs, missing one or more modalities is a typical concern of multimodal methods. We hence propose a novel deep learning-based classification framework where generative module employing Cycle GANs was adopted to impute missing data within the latent space. Additionally, we adopted an Explainable AI method, Integrated Gradients, to extract input features relevance, enhancing our understanding of the learned representations. Two critical tasks were addressed: AD detection and MCI conversion prediction. Experimental results showed that our model was able to reach the SOA in the classification of CN/AD reaching an average test accuracy of $0.926pm0.02$. For the MCI task, we achieved an average prediction accuracy of $0.711pm0.01$ using the pre-trained model for CN/AD. The interpretability analysis revealed significant grey matter modulations in cortical and subcortical brain areas well known for their association with AD. Moreover, impairments in sensory-motor and visual resting state network connectivity along the disease continuum, as well as mutations in SNPs defining biological processes linked to amyloid-beta and cholesterol formation clearance and regulation, were identified as contributors to the achieved performance. Overall, our integrative deep learning approach shows promise for AD detection and MCI prediction, while shading light on important biological insights.

Read more

6/21/2024

Multimodal MRI-based Detection of Amyloid Status in Alzheimer's Disease Continuum

Multimodal MRI-based Detection of Amyloid Status in Alzheimer's Disease Continuum

Giorgio Dolci (Department of Computer Science, University of Verona, Verona, Italy, Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy, Tri-Institutional Center for Translational Research in Neuroimaging and Data Science), Charles A. Ellis (Tri-Institutional Center for Translational Research in Neuroimaging and Data Science), Federica Cruciani (Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy), Lorenza Brusini (Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy), Anees Abrol (Tri-Institutional Center for Translational Research in Neuroimaging and Data Science), Ilaria Boscolo Galazzo (Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy), Gloria Menegaz (Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy), Vince D. Calhoun (Tri-Institutional Center for Translational Research in Neuroimaging and Data Science)

YC

0

Reddit

0

Amyloid-$beta$ (A$beta$) plaques in conjunction with hyperphosphorylated tau proteins in the form of neurofibrillary tangles are the two neuropathological hallmarks of Alzheimer's disease (AD). In particular, the accumulation of A$beta$ plaques, as evinced by the A/T/N (amyloid/tau/neurodegeneration) framework, marks the initial stage. Thus, the identification of individuals with A$beta$ positivity could enable early diagnosis and potentially lead to more effective interventions. Deep learning methods relying mainly on amyloid PET images have been employed to this end. However, PET imaging has some disadvantages, including the need of radiotracers and expensive acquisitions. Hence, in this work, we propose a novel multimodal approach that integrates information from structural, functional, and diffusion MRI data to discriminate A$beta$ status in the AD continuum. Our method achieved an accuracy of $0.762pm0.04$. Furthermore, a textit{post-hoc} explainability analysis (guided backpropagation) was performed to retrieve the brain regions that most influenced the model predictions. This analysis identified some key regions that were common across modalities, some of which were well-established AD-discriminative biomarkers and related to A$beta$ deposition, such as the hippocampus, thalamus, precuneus, and cingulate gyrus. Hence, our study demonstrates the potential viability of MRI-based characterization of A$beta$ status, paving the way for further research in this domain.

Read more

6/21/2024

Trustworthy Enhanced Multi-view Multi-modal Alzheimer's Disease Prediction with Brain-wide Imaging Transcriptomics Data

Trustworthy Enhanced Multi-view Multi-modal Alzheimer's Disease Prediction with Brain-wide Imaging Transcriptomics Data

Shan Cong, Zhoujie Fan, Hongwei Liu, Yinghan Zhang, Xin Wang, Haoran Luo, Xiaohui Yao

YC

0

Reddit

0

Brain transcriptomics provides insights into the molecular mechanisms by which the brain coordinates its functions and processes. However, existing multimodal methods for predicting Alzheimer's disease (AD) primarily rely on imaging and sometimes genetic data, often neglecting the transcriptomic basis of brain. Furthermore, while striving to integrate complementary information between modalities, most studies overlook the informativeness disparities between modalities. Here, we propose TMM, a trusted multiview multimodal graph attention framework for AD diagnosis, using extensive brain-wide transcriptomics and imaging data. First, we construct view-specific brain regional co-function networks (RRIs) from transcriptomics and multimodal radiomics data to incorporate interaction information from both biomolecular and imaging perspectives. Next, we apply graph attention (GAT) processing to each RRI network to produce graph embeddings and employ cross-modal attention to fuse transcriptomics-derived embedding with each imagingderived embedding. Finally, a novel true-false-harmonized class probability (TFCP) strategy is designed to assess and adaptively adjust the prediction confidence of each modality for AD diagnosis. We evaluate TMM using the AHBA database with brain-wide transcriptomics data and the ADNI database with three imaging modalities (AV45-PET, FDG-PET, and VBM-MRI). The results demonstrate the superiority of our method in identifying AD, EMCI, and LMCI compared to state-of-the-arts. Code and data are available at https://github.com/Yaolab-fantastic/TMM.

Read more

6/24/2024

Cross-Modality Translation with Generative Adversarial Networks to Unveil Alzheimer's Disease Biomarkers

Cross-Modality Translation with Generative Adversarial Networks to Unveil Alzheimer's Disease Biomarkers

Reihaneh Hassanzadeh, Anees Abrol, Hamid Reza Hassanzadeh, Vince D. Calhoun

YC

0

Reddit

0

Generative approaches for cross-modality transformation have recently gained significant attention in neuroimaging. While most previous work has focused on case-control data, the application of generative models to disorder-specific datasets and their ability to preserve diagnostic patterns remain relatively unexplored. Hence, in this study, we investigated the use of a generative adversarial network (GAN) in the context of Alzheimer's disease (AD) to generate functional network connectivity (FNC) and T1-weighted structural magnetic resonance imaging data from each other. We employed a cycle-GAN to synthesize data in an unpaired data transition and enhanced the transition by integrating weak supervision in cases where paired data were available. Our findings revealed that our model could offer remarkable capability, achieving a structural similarity index measure (SSIM) of $0.89 pm 0.003$ for T1s and a correlation of $0.71 pm 0.004$ for FNCs. Moreover, our qualitative analysis revealed similar patterns between generated and actual data when comparing AD to cognitively normal (CN) individuals. In particular, we observed significantly increased functional connectivity in cerebellar-sensory motor and cerebellar-visual networks and reduced connectivity in cerebellar-subcortical, auditory-sensory motor, sensory motor-visual, and cerebellar-cognitive control networks. Additionally, the T1 images generated by our model showed a similar pattern of atrophy in the hippocampal and other temporal regions of Alzheimer's patients.

Read more

5/10/2024