705 research outputs found

    Artificial intelligence for dementia research methods optimization

    Get PDF
    Artificial intelligence (AI) and machine learning (ML) approaches are increasingly being used in dementia research. However, several methodological challenges exist that may limit the insights we can obtain from high-dimensional data and our ability to translate these findings into improved patient outcomes. To improve reproducibility and replicability, researchers should make their well-documented code and modeling pipelines openly available. Data should also be shared where appropriate. To enhance the acceptability of models and AI-enabled systems to users, researchers should prioritize interpretable methods that provide insights into how decisions are generated. Models should be developed using multiple, diverse datasets to improve robustness, generalizability, and reduce potentially harmful bias. To improve clarity and reproducibility, researchers should adhere to reporting guidelines that are co-produced with multiple stakeholders. If these methodological challenges are overcome, AI and ML hold enormous promise for changing the landscape of dementia research and care. HIGHLIGHTS: Machine learning (ML) can improve diagnosis, prevention, and management of dementia. Inadequate reporting of ML procedures affects reproduction/replication of results. ML models built on unrepresentative datasets do not generalize to new datasets. Obligatory metrics for certain model structures and use cases have not been defined. Interpretability and trust in ML predictions are barriers to clinical translation

    Predictive analytics applied to Alzheimerโ€™s disease : a data visualisation framework for understanding current research and future challenges

    Get PDF
    Dissertation as a partial requirement for obtaining a masterโ€™s degree in information management, with a specialisation in Business Intelligence and Knowledge Management.Big Data is, nowadays, regarded as a tool for improving the healthcare sector in many areas, such as in its economic side, by trying to search for operational efficiency gaps, and in personalised treatment, by selecting the best drug for the patient, for instance. Data science can play a key role in identifying diseases in an early stage, or even when there are no signs of it, track its progress, quickly identify the efficacy of treatments and suggest alternative ones. Therefore, the prevention side of healthcare can be enhanced with the usage of state-of-the-art predictive big data analytics and machine learning methods, integrating the available, complex, heterogeneous, yet sparse, data from multiple sources, towards a better disease and pathology patterns identification. It can be applied for the diagnostic challenging neurodegenerative disorders; the identification of the patterns that trigger those disorders can make possible to identify more risk factors, biomarkers, in every human being. With that, we can improve the effectiveness of the medical interventions, helping people to stay healthy and active for a longer period. In this work, a review of the state of science about predictive big data analytics is done, concerning its application to Alzheimerโ€™s Disease early diagnosis. It is done by searching and summarising the scientific articles published in respectable online sources, putting together all the information that is spread out in the world wide web, with the goal of enhancing knowledge management and collaboration practices about the topic. Furthermore, an interactive data visualisation tool to better manage and identify the scientific articles is develop, delivering, in this way, a holistic visual overview of the developments done in the important field of Alzheimerโ€™s Disease diagnosis.Big Data รฉ hoje considerada uma ferramenta para melhorar o sector da saรบde em muitas รกreas, tais como na sua vertente mais econรณmica, tentando encontrar lacunas de eficiรชncia operacional, e no tratamento personalizado, selecionando o melhor medicamento para o paciente, por exemplo. A ciรชncia de dados pode desempenhar um papel fundamental na identificaรงรฃo de doenรงas em um estรกgio inicial, ou mesmo quando nรฃo hรก sinais dela, acompanhar o seu progresso, identificar rapidamente a eficรกcia dos tratamentos indicados ao paciente e sugerir alternativas. Portanto, o lado preventivo dos cuidados de saรบde pode ser bastante melhorado com o uso de mรฉtodos avanรงados de anรกlise preditiva com big data e de machine learning, integrando os dados disponรญveis, geralmente complexos, heterogรฉneos e esparsos provenientes de mรบltiplas fontes, para uma melhor identificaรงรฃo de padrรตes patolรณgicos e da doenรงa. Estes mรฉtodos podem ser aplicados nas doenรงas neurodegenerativas que ainda sรฃo um grande desafio no seu diagnรณstico; a identificaรงรฃo dos padrรตes que desencadeiam esses distรบrbios pode possibilitar a identificaรงรฃo de mais fatores de risco, biomarcadores, em todo e qualquer ser humano. Com isso, podemos melhorar a eficรกcia das intervenรงรตes mรฉdicas, ajudando as pessoas a permanecerem saudรกveis e ativas por um perรญodo mais longo. Neste trabalho, รฉ feita uma revisรฃo do estado da arte sobre a anรกlise preditiva com big data, no que diz respeito ร  sua aplicaรงรฃo ao diagnรณstico precoce da Doenรงa de Alzheimer. Isto foi realizado atravรฉs da pesquisa exaustiva e resumo de um grande nรบmero de artigos cientรญficos publicados em fontes online de referรชncia na รกrea, reunindo a informaรงรฃo que estรก amplamente espalhada na world wide web, com o objetivo de aprimorar a gestรฃo do conhecimento e as prรกticas de colaboraรงรฃo sobre o tema. Alรฉm disso, uma ferramenta interativa de visualizaรงรฃo de dados para melhor gerir e identificar os artigos cientรญficos foi desenvolvida, fornecendo, desta forma, uma visรฃo holรญstica dos avanรงos cientรญfico feitos no importante campo do diagnรณstico da Doenรงa de Alzheimer

    An MRI-Derived Definition of MCI-to-AD Conversion for Long-Term, Automati c Prognosis of MCI Patients

    Get PDF
    Alzheimer's disease (AD) and mild cognitive impairment (MCI), continue to be widely studied. While there is no consensus on whether MCIs actually "convert" to AD, the more important question is not whether MCIs convert, but what is the best such definition. We focus on automatic prognostication, nominally using only a baseline image brain scan, of whether an MCI individual will convert to AD within a multi-year period following the initial clinical visit. This is in fact not a traditional supervised learning problem since, in ADNI, there are no definitive labeled examples of MCI conversion. Prior works have defined MCI subclasses based on whether or not clinical/cognitive scores such as CDR significantly change from baseline. There are concerns with these definitions, however, since e.g. most MCIs (and ADs) do not change from a baseline CDR=0.5, even while physiological changes may be occurring. These works ignore rich phenotypical information in an MCI patient's brain scan and labeled AD and Control examples, in defining conversion. We propose an innovative conversion definition, wherein an MCI patient is declared to be a converter if any of the patient's brain scans (at follow-up visits) are classified "AD" by an (accurately-designed) Control-AD classifier. This novel definition bootstraps the design of a second classifier, specifically trained to predict whether or not MCIs will convert. This second classifier thus predicts whether an AD-Control classifier will predict that a patient has AD. Our results demonstrate this new definition leads not only to much higher prognostic accuracy than by-CDR conversion, but also to subpopulations much more consistent with known AD brain region biomarkers. We also identify key prognostic region biomarkers, essential for accurately discriminating the converter and nonconverter groups

    ๋”ฅ๋Ÿฌ๋‹ ๊ธฐ๋ฐ˜ ๊ตฐ์ง‘ํ™” ๋ฐฉ๋ฒ•์„ ์ด์šฉํ•˜์—ฌ FDG PET์—์„œ ์•Œ์ธ ํ•˜์ด๋จธ๋ณ‘์˜ ๊ณต๊ฐ„์  ๋‡Œ ๋Œ€์‚ฌ ํŒจํ„ด์˜ ํŠน์ง•์  ์•„ํ˜• ๋ถ„๋ฅ˜

    Get PDF
    ํ•™์œ„๋…ผ๋ฌธ(๋ฐ•์‚ฌ) -- ์„œ์šธ๋Œ€ํ•™๊ต๋Œ€ํ•™์› : ์œตํ•ฉ๊ณผํ•™๊ธฐ์ˆ ๋Œ€ํ•™์› ๋ถ„์ž์˜ํ•™ ๋ฐ ๋ฐ”์ด์˜ค์ œ์•ฝํ•™๊ณผ, 2022.2. ์ด๋™์ˆ˜.์•Œ์ธ ํ•˜์ด๋จธ๋ณ‘์€ ์•„๋ฐ€๋กœ์ด๋“œ์™€ ํƒ€์šฐ ์นจ์ฐฉ๊ณผ ๊ฐ™์€ ๋ณ‘๋ฆฌํ•™์  ํŠน์ง•์„ ๊ณต์œ ํ•จ์—๋„ ๋ถˆ๊ตฌํ•˜๊ณ  ๊ด‘๋ฒ”์œ„ํ•œ ์ž„์ƒ๋ณ‘๋ฆฌํ•™์  ํŠน์„ฑ์„ ๋ณด์ธ๋‹ค. ๋ณธ ์—ฐ๊ตฌ์—์„œ๋Š” ๋”ฅ๋Ÿฌ๋‹ ๊ธฐ๋ฐ˜ ๊ตฐ์ง‘ํ™” ๋ฐฉ๋ฒ•์„ ์ด์šฉํ•˜์—ฌ FDG PET ์˜์ƒ์—์„œ ์•Œ์ธ ํ•˜์ด๋จธ๋ณ‘ ํŠน์ง•์  ์•„ํ˜•์„ ๋ถ„๋ฅ˜ํ•˜์—ฌ ์‹ ๊ฒฝ ํ‡ดํ–‰์˜ ๊ณต๊ฐ„์  ๋‡Œ ๋Œ€์‚ฌ ํŒจํ„ด์„ ์ดํ•ดํ•˜๊ณ ์ž ํ•˜์˜€์œผ๋ฉฐ, ๊ณต๊ฐ„์  ๋‡Œ ๋Œ€์‚ฌ ํŒจํ„ด์— ์˜ํ•ด ์ •์˜๋œ ์•„ํ˜•์˜ ์ž„์ƒ๋ณ‘๋ฆฌํ•™์  ํŠน์ง•์„ ๋ฐํžˆ๊ณ ์ž ํ•˜์˜€๋‹ค. Alzheimerโ€™s Disease Neuroimaging Initiative(ADNI) ๋ฐ์ดํ„ฐ๋ฒ ์ด์Šค๋กœ๋ถ€ํ„ฐ ์ฒซ๋ฒˆ์งธ ๋ฐฉ๋ฌธ ๋ฐ ์ถ”์  ๋ฐฉ๋ฌธ์„ ํฌํ•จํ•œ ์•Œ์ธ ํ•˜์ด๋จธ๋ณ‘, ๊ฒฝ๋„์ธ์ง€์žฅ์• , ์ธ์ง€ ์ •์ƒ๊ตฐ์˜ ์ด 3620๊ฐœ์˜ FDG ๋‡Œ ์–‘์ „์ž๋‹จ์ธต์ดฌ์˜(PET) ์˜์ƒ์„ ์ˆ˜์ง‘ํ•˜์˜€๋‹ค. ์•Œ์ธ ํ•˜์ด๋จธ๋ณ‘์—์„œ ์งˆ๋ณ‘์˜ ์ง„ํ–‰ ์™ธ์˜ ๋‡Œ ๋Œ€์‚ฌ ํŒจํ„ด์„ ๋‚˜ํƒ€๋‚ด๋Š” ํ‘œํ˜„(representation)์„ ์ฐพ๊ธฐ ์œ„ํ•˜์—ฌ, ์กฐ๊ฑด๋ถ€ ๋ณ€์ดํ˜• ์˜คํ† ์ธ์ฝ”๋”(conditional variational autoencoder)๋ฅผ ์‚ฌ์šฉํ•˜์˜€์œผ๋ฉฐ, ์ธ์ฝ”๋”ฉ๋œ ํ‘œํ˜„์œผ๋กœ๋ถ€ํ„ฐ ๊ตฐ์ง‘ํ™”(clustering)๋ฅผ ์‹œํ–‰ํ•˜์˜€๋‹ค. ์•Œ์ธ ํ•˜์ด๋จธ๋ณ‘์˜ ๋‡Œ FDG PET (n=838)๊ณผ CDR-SB(Clinical Demetria Rating Scale Sum of Boxes) ์ ์ˆ˜๊ฐ€ cVAE ๋ชจ๋ธ์˜ ์ž…๋ ฅ๊ฐ’์œผ๋กœ ์‚ฌ์šฉ๋˜์—ˆ์œผ๋ฉฐ, ๊ตฐ์ง‘ํ™”์—๋Š” k-means ์•Œ๊ณ ๋ฆฌ์ฆ˜์ด ์‚ฌ์šฉ๋˜์—ˆ๋‹ค. ํ›ˆ๋ จ๋œ ๋”ฅ๋Ÿฌ๋‹ ๋ชจ๋ธ์€ ๊ฒฝ๋„์ธ์ง€์žฅ์• ๊ตฐ (n=1761)์˜ ๋‡Œ FDG PET์— ์ „์ด(transfer)๋˜์–ด ๊ฐ ์•„ํ˜•์˜ ์„œ๋กœ ๋‹ค๋ฅธ ๊ถค์ (trajectory)๊ณผ ์˜ˆํ›„๋ฅผ ๋ฐํžˆ๊ณ ์ž ํ•˜์˜€๋‹ค. ํ†ต๊ณ„์  ํŒŒ๋ผ๋ฏธํ„ฐ ์ง€๋„์ž‘์„ฑ๋ฒ•(Statistical Parametric Mapping, SPM)์„ ์ด์šฉํ•˜์—ฌ ๊ฐ ๊ตฐ์ง‘์˜ ๊ณต๊ฐ„์  ํŒจํ„ด์„ ์‹œ๊ฐํ™” ํ•˜์˜€์œผ๋ฉฐ, ๊ฐ ๊ตฐ์ง‘์˜ ์ž„์ƒ์  ๋ฐ ์ƒ๋ฌผํ•™์  ํŠน์ง•์„ ๋น„๊ตํ•˜์˜€๋‹ค. ๋˜ํ•œ ์•„ํ˜• ๋ณ„ ๊ฒฝ๋„์ธ์ง€์žฅ์• ๋กœ๋ถ€ํ„ฐ ์•Œ์ธ ํ•˜์ด๋จธ๋ณ‘์œผ๋กœ ์ „ํ™˜๋˜๋Š” ๋น„์œจ์„ ๋น„๊ตํ•˜์˜€๋‹ค. ๋”ฅ๋Ÿฌ๋‹ ๊ธฐ๋ฐ˜ ๊ตฐ์ง‘ํ™” ๋ฐฉ๋ฒ•์œผ๋กœ 4๊ฐœ์˜ ํŠน์ง•์  ์•„ํ˜•์ด ๋ถ„๋ฅ˜๋˜์—ˆ๋‹ค. (i) S1 (angular): ๋ชจ์ด๋ž‘(angular gyrus)์—์„œ ํ˜„์ €ํ•œ ๋Œ€์‚ฌ ์ €ํ•˜๋ฅผ ๋ณด์ด๋ฉฐ ๋ถ„์‚ฐ๋œ ํ”ผ์งˆ์˜ ๋Œ€์‚ฌ ์ €ํ•˜ ํŒจํ„ด, ๋‚จ์„ฑ์—์„œ ๋นˆ๋„ ๋†’์Œ, ๋” ๋งŽ์€ ์•„๋ฐ€๋กœ์ด๋“œ ์นจ์ฐฉ, ๋” ์ ์€ ํƒ€์šฐ ์นจ์ฐฉ, ๋” ์‹ฌํ•œ ํ•ด๋งˆ ์œ„์ถ•, ์ดˆ๊ธฐ ๋‹จ๊ณ„์˜ ์ธ์ง€ ์ €ํ•˜์˜ ํŠน์ง•์„ ๋ณด์˜€๋‹ค. (ii) S2 (occipital): ํ›„๋‘์—ฝ(occipital) ํ”ผ์งˆ์—์„œ ํ˜„์ €ํ•œ ๋Œ€์‚ฌ ์ €ํ•˜๋ฅผ ๋ณด์ด๋ฉฐ ํ›„๋ถ€ ์šฐ์„ธํ•œ ๋Œ€์‚ฌ ์ €ํ•˜ ํŒจํ„ด, ๋” ์ ์€ ์—ฐ๋ น, ๋” ๋งŽ์€ ํƒ€์šฐ, ๋” ์ ์€ ํ•ด๋งˆ ์œ„์ถ•, ๋” ๋‚ฎ์€ ์ง‘ํ–‰ ๋ฐ ์‹œ๊ณต๊ฐ„ ์ ์ˆ˜, ๊ฒฝ๋„์ธ์ง€์žฅ์• ๋กœ๋ถ€ํ„ฐ ์•Œ์ธ ํ•˜์ด๋จธ๋ณ‘์œผ๋กœ์˜ ๋น ๋ฅธ ์ „ํ™˜์˜ ํŠน์ง•์„ ๋ณด์˜€๋‹ค. (iii) S3(orbitofrontal): ์•ˆ์™€์ „๋‘(orbitofrontal) ํ”ผ์งˆ์—์„œ ํ˜„์ €ํ•œ ๋Œ€์‚ฌ ์ €ํ•˜๋ฅผ ๋ณด์ด๋ฉฐ ์ „๋ฐฉ ์šฐ์„ธํ•œ ๋Œ€์‚ฌ ์ €ํ•˜ ํŒจํ„ด, ๋” ๋†’์€ ์—ฐ๋ น, ๋” ์ ์€ ์•„๋ฐ€๋กœ์ด๋“œ ์นจ์ฐฉ, ๋” ์‹ฌํ•œ ํ•ด๋งˆ ์œ„์ถ•, ๋” ๋†’์€ ์ง‘ํ–‰ ๋ฐ ์‹œ๊ณต๊ฐ„ ์ ์ˆ˜์˜ ํŠน์ง•์„ ๋ณด์˜€๋‹ค. (iv) S4(minimal): ์ตœ์†Œ์˜ ๋Œ€์‚ฌ ์ €ํ•˜๋ฅผ ๋ณด์ž„, ์—ฌ์„ฑ์—์„œ ๋นˆ๋„ ๋†’์Œ, ๋” ์ ์€ ์•„๋ฐ€๋กœ์ด๋“œ ์นจ์ฐฉ, ๋” ๋งŽ์€ ํƒ€์šฐ ์นจ์ฐฉ, ๋” ์ ์€ ํ•ด๋งˆ ์œ„์ถ•, ๋” ๋†’์€ ์ธ์ง€๊ธฐ๋Šฅ ์ ์ˆ˜์˜ ํŠน์ง•์„ ๋ณด์˜€๋‹ค. ๊ฒฐ๋ก ์ ์œผ๋กœ, ๋ณธ ์—ฐ๊ตฌ์—์„œ ์šฐ๋ฆฌ๋Š” ์„œ๋กœ ๋‹ค๋ฅธ ๋‡Œ ๋ณ‘๋ฆฌ ๋ฐ ์ž„์ƒ ํŠน์„ฑ์„ ๊ฐ€์ง„ ์•Œ์ธ ํ•˜์ด๋จธ๋ณ‘์˜ ํŠน์ง•์  ์•„ํ˜•์„ ๋ถ„๋ฅ˜ํ•˜์˜€๋‹ค. ๋˜ํ•œ ์šฐ๋ฆฌ ๋”ฅ๋Ÿฌ๋‹ ๋ชจ๋ธ์€ ๊ฒฝ๋„์ธ์ง€์žฅ์• ๊ตฐ์— ์„ฑ๊ณต์ ์œผ๋กœ ์ „์ด๋˜์–ด ์•„ํ˜• ๋ณ„ ๊ฒฝ๋„์ธ์ง€์žฅ์• ๋กœ๋ถ€ํ„ฐ ์•Œ์ธ ํ•˜์ด๋จธ๋ณ‘์œผ๋กœ ์ „ํ™˜๋˜๋Š” ์˜ˆํ›„๋ฅผ ์˜ˆ์ธกํ•  ์ˆ˜ ์žˆ์—ˆ๋‹ค. ๋ณธ ๊ฒฐ๊ณผ๋Š” FDG PET์—์„œ ์•Œ์ธ ํ•˜์ด๋จธ๋ณ‘์˜ ํŠน์ง•์  ์•„ํ˜•์€ ๊ฐœ์ธ์˜ ์ž„์ƒ ๊ฒฐ๊ณผ์— ์˜ํ–ฅ์„ ๋ฏธ์น  ์ˆ˜ ์žˆ๊ณ , ๋ณ‘ํƒœ์ƒ๋ฆฌํ•™ ์ธก๋ฉด์—์„œ ์•Œ์ธ ํ•˜์ด๋จธ๋ณ‘์˜ ๊ด‘๋ฒ”์œ„ํ•œ ์ŠคํŽ™ํŠธ๋Ÿผ์„ ์ดํ•ดํ•˜๋Š”๋ฐ ๋‹จ์„œ๋ฅผ ์ œ๊ณตํ•  ์ˆ˜ ์žˆ์Œ์„ ์‹œ์‚ฌํ•œ๋‹ค.Alzheimerโ€™s disease (AD) presents a broad spectrum of clinicopathologic profiles, despite common pathologic features including amyloid and tau deposition. Here, we aimed to identify AD subtypes using deep learning-based clustering on FDG PET images to understand distinct spatial patterns of neurodegeneration. We also aimed to investigate clinicopathologic features of subtypes defined by spatial brain metabolism patterns. A total of 3620 FDG brain PET images with AD, mild cognitive impairment (MCI), and cognitively normal controls (CN) at baseline and follow-up visits were obtained from Alzheimerโ€™s Disease Neuroimaging Initiative (ADNI) database. In order to identify representations of brain metabolism patterns different from disease progression in AD, a conditional variational autoencoder (cVAE) was used, followed by clustering using the encoded representations. FDG brain PET images with AD (n=838) and Clinical Demetria Rating Scale Sum of Boxes (CDR-SB) scores were used as inputs of cVAE model and the k-means algorithm was applied for the clustering. The trained deep learning model was also transferred to FDG brain PET image with MCI (n=1761) to identify differential trajectories and prognosis of subtypes. Statistical parametric maps were generated to visualize spatial patterns of clusters, and clinical and biological characteristics were compared among the clusters. The conversion rate from MCI to AD was also compared among the subtypes. Four distinct subtypes were identified by deep learning-based FDG PET clusters: (i) S1 (angular), showing prominent hypometabolism in the angular gyrus with a diffuse cortical hypometabolism pattern; frequent in males; more amyloid; less tau; more hippocampal atrophy; cognitive decline in the earlier stage. (ii) S2 (occipital), showing prominent hypometabolism in the occipital cortex with a posterior-predominant hypometabolism pattern; younger age; more tau; less hippocampal atrophy; lower executive and visuospatial scores; faster conversion from MCI to AD. (iii) S3 (orbitofrontal), showing prominent hypometabolism in the orbitofrontal cortex with an anterior-predominant hypometabolism pattern; older age; less amyloid; more hippocampal atrophy; higher executive and visuospatial scores. (iv) S4 (minimal), showing minimal hypometabolism; frequent in females; less amyloid; more tau; less hippocampal atrophy; higher cognitive scores. In conclusion, we could identify distinct subtypes in AD with different brain pathologies and clinical profiles. Also, our deep learning model was successfully transferred to MCI to predict the prognosis of subtypes for conversion from MCI to AD. Our results suggest that distinct AD subtypes on FDG PET may have implications for the individual clinical outcomes and provide a clue to understanding a broad spectrum of AD in terms of pathophysiology.1. Introduction 1 1.1 Heterogeneity of Alzheimer's disease 1 1.2 FDG PET as a biomarker of Alzheimer's disease 1 1.3 Biologic subtypes of Alzheimer's disease 2 1.4 Dimensionality reduction methods 5 1.5 Variational autoencoder for clustering 8 1.6 Final goal of the study 10 2. Methods 11 2.1 Subjects 11 2.2 FDG PET data acquisition and preprocessing 12 2.3 Deep learning-based model for representations of FDG PET in AD 12 2.4 Clustering method for AD subtypes on FDG PET 17 2.5 Transfer of deep learning-based FDG PET cluster model for MCI subtypes 17 2.6 Visualization of subtype-specific spatial brain metabolism pattern 21 2.7 Clinical and biological characterization 21 2.8 Prognosis prediction of MCI subtypes 22 2.9 Generation of subtype-specific FDG PET images 22 2.10 Statistical analysis 23 3. Results 24 3.1 Deep learning-based FDG PET clusters 24 3.2 Spatial brain metabolism pattern in AD subtypes 27 3.3 Clinical and biological characterization in AD subtypes 32 3.4 Subtype-specific spatial metabolism patterns resemble in MCI 43 3.5 Clinical and biological characterization in MCI subtypes 50 3.6 Prognosis prediction of subtypes for conversion from MCI to AD 56 3.7 Generating FDG PET images of AD subtypes 61 4. Discussion 66 4.1 Limitations of previous subtyping approach 68 4.2 Interpretation of results 68 4.3 Strength of our deep learning-based clustering approach 73 4.4 Strength of our deep learning-based AD subtypes 77 4.5 Limitations and future directions 82 5. Conclusion 83 References 84 Supplementary Figures 99 ๊ตญ๋ฌธ ์ดˆ๋ก 101๋ฐ•

    Artificial intelligence for diagnostic and prognostic neuroimaging in dementia: a systematic review

    Get PDF
    Introduction: Artificial intelligence (AI) and neuroimaging offer new opportunities for diagnosis and prognosis of dementia. Methods: We systematically reviewed studies reporting AI for neuroimaging in diagnosis and/or prognosis of cognitive neurodegenerative diseases. Results: A total of 255 studies were identified. Most studies relied on the Alzheimer's Disease Neuroimaging Initiative dataset. Algorithmic classifiers were the most commonly used AI method (48%) and discriminative models performed best for differentiating Alzheimer's disease from controls. The accuracy of algorithms varied with the patient cohort, imaging modalities, and stratifiers used. Few studies performed validation in an independent cohort. Discussion: The literature has several methodological limitations including lack of sufficient algorithm development descriptions and standard definitions. We make recommendations to improve model validation including addressing key clinical questions, providing sufficient description of AI methods and validating findings in independent datasets. Collaborative approaches between experts in AI and medicine will help achieve the promising potential of AI tools in practice. Highlights: There has been a rapid expansion in the use of machine learning for diagnosis and prognosis in neurodegenerative disease Most studies (71%) relied on the Alzheimer's Disease Neuroimaging Initiative (ADNI) dataset with no other individual dataset used more than five times There has been a recent rise in the use of more complex discriminative models (e.g., neural networks) that performed better than other classifiers for classification of AD vs healthy controls We make recommendations to address methodological considerations, addressing key clinical questions, and validation We also make recommendations for the field more broadly to standardize outcome measures, address gaps in the literature, and monitor sources of bias
    • โ€ฆ
    corecore