1
|
Kaisaridi S, Herve D, Jabouley A, Reyes S, Machado C, Guey S, Taleb A, Fernandes F, Chabriat H, Tezenas Du Montcel S. Determining Clinical Disease Progression in Symptomatic Patients With CADASIL. Neurology 2025; 104:e210193. [PMID: 39689282 DOI: 10.1212/wnl.0000000000210193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 10/22/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most frequent small artery brain disease caused by pathogenic variants of the NOTCH3 gene. During the disease, we still do not know how the various deficits progress and develop with each other at different stages of the disease. We aim to model disease progression and identify possible progressive subgroups and the effects of different covariates on clinical worsening. METHODS Data were obtained from patients followed in the French CADASIL referral center, who were aged 25-80 years and had completed at least 2 visits and one of 14 clinical scores. Progression and variability were assessed using a disease course model (Leaspy). A Gaussian mixture model was used to identify different progression subgroups. Logistic regressions were used to compare the characteristics between groups. RESULTS In 395 patients along 2,007 visits, the follow-up ranged from 6 months to 19 years, with a mean of 7.5 years. They were 45% men with a mean age of 52.2 years. The evolution curves of the different scores showed that clinical manifestations develop heterogeneously and can vary considerably depending on the disease stage. We identified an early-onset, rapidly progressing subgroup of patients with earlier motor symptoms and focal neurologic deficits (median time shift 59 [Q1-Q3 48.9-66.3], median acceleration rate 0.84 [0.07-1.31]) and a late-onset slowly progressing group with earlier cognitive symptoms (median time shift 69.2 [63.4-75.1], median acceleration rate -0.18 [-0.48 to 0.14]). Male sex, lower education level, hypertension, and NOTCH3 pathogenic variant location within epidermal growth factor-like repeat (EGFr) 1-6 were found to be associated with this group difference. DISCUSSION Our results suggest a gradual and heterogeneous decline in different clinical and cognitive performances over the lifetime of patients with CADASIL. Two progression profiles-one rapid and early and the other, more delayed and slower-are possible after the onset of symptoms. A major limitation of our study is that the clusters were assessed post hoc, which may induce some bias. Overall, male sex, low level of education, pathogenic variant location in EGFr 1 to 6 domains, smoking, and/or arterial hypertension may affect the clinical progression of the disease.
Collapse
Affiliation(s)
- Sofia Kaisaridi
- From the ARAMIS (S.K., S.T.D.M.), Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Groupe Hospitalier Sorbonne Université; Centre de référence pour les maladies vasculaires rares du cerveau et de l'œil (CERVCO) and Centre Neurovascular Translationnel (CNVT) (D.H., A.J., S.R., C.M., S.G., A.T., F.F., H.C.), AP-HP, Paris; and INSERM U1141 - FHU NeuroVasc (D.H., S.G., H.C.), Université Paris Cité, France
| | - Dominique Herve
- From the ARAMIS (S.K., S.T.D.M.), Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Groupe Hospitalier Sorbonne Université; Centre de référence pour les maladies vasculaires rares du cerveau et de l'œil (CERVCO) and Centre Neurovascular Translationnel (CNVT) (D.H., A.J., S.R., C.M., S.G., A.T., F.F., H.C.), AP-HP, Paris; and INSERM U1141 - FHU NeuroVasc (D.H., S.G., H.C.), Université Paris Cité, France
| | - Aude Jabouley
- From the ARAMIS (S.K., S.T.D.M.), Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Groupe Hospitalier Sorbonne Université; Centre de référence pour les maladies vasculaires rares du cerveau et de l'œil (CERVCO) and Centre Neurovascular Translationnel (CNVT) (D.H., A.J., S.R., C.M., S.G., A.T., F.F., H.C.), AP-HP, Paris; and INSERM U1141 - FHU NeuroVasc (D.H., S.G., H.C.), Université Paris Cité, France
| | - Sonia Reyes
- From the ARAMIS (S.K., S.T.D.M.), Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Groupe Hospitalier Sorbonne Université; Centre de référence pour les maladies vasculaires rares du cerveau et de l'œil (CERVCO) and Centre Neurovascular Translationnel (CNVT) (D.H., A.J., S.R., C.M., S.G., A.T., F.F., H.C.), AP-HP, Paris; and INSERM U1141 - FHU NeuroVasc (D.H., S.G., H.C.), Université Paris Cité, France
| | - Carla Machado
- From the ARAMIS (S.K., S.T.D.M.), Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Groupe Hospitalier Sorbonne Université; Centre de référence pour les maladies vasculaires rares du cerveau et de l'œil (CERVCO) and Centre Neurovascular Translationnel (CNVT) (D.H., A.J., S.R., C.M., S.G., A.T., F.F., H.C.), AP-HP, Paris; and INSERM U1141 - FHU NeuroVasc (D.H., S.G., H.C.), Université Paris Cité, France
| | - Stéphanie Guey
- From the ARAMIS (S.K., S.T.D.M.), Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Groupe Hospitalier Sorbonne Université; Centre de référence pour les maladies vasculaires rares du cerveau et de l'œil (CERVCO) and Centre Neurovascular Translationnel (CNVT) (D.H., A.J., S.R., C.M., S.G., A.T., F.F., H.C.), AP-HP, Paris; and INSERM U1141 - FHU NeuroVasc (D.H., S.G., H.C.), Université Paris Cité, France
| | - Abbas Taleb
- From the ARAMIS (S.K., S.T.D.M.), Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Groupe Hospitalier Sorbonne Université; Centre de référence pour les maladies vasculaires rares du cerveau et de l'œil (CERVCO) and Centre Neurovascular Translationnel (CNVT) (D.H., A.J., S.R., C.M., S.G., A.T., F.F., H.C.), AP-HP, Paris; and INSERM U1141 - FHU NeuroVasc (D.H., S.G., H.C.), Université Paris Cité, France
| | - Fanny Fernandes
- From the ARAMIS (S.K., S.T.D.M.), Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Groupe Hospitalier Sorbonne Université; Centre de référence pour les maladies vasculaires rares du cerveau et de l'œil (CERVCO) and Centre Neurovascular Translationnel (CNVT) (D.H., A.J., S.R., C.M., S.G., A.T., F.F., H.C.), AP-HP, Paris; and INSERM U1141 - FHU NeuroVasc (D.H., S.G., H.C.), Université Paris Cité, France
| | - Hugues Chabriat
- From the ARAMIS (S.K., S.T.D.M.), Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Groupe Hospitalier Sorbonne Université; Centre de référence pour les maladies vasculaires rares du cerveau et de l'œil (CERVCO) and Centre Neurovascular Translationnel (CNVT) (D.H., A.J., S.R., C.M., S.G., A.T., F.F., H.C.), AP-HP, Paris; and INSERM U1141 - FHU NeuroVasc (D.H., S.G., H.C.), Université Paris Cité, France
| | - Sophie Tezenas Du Montcel
- From the ARAMIS (S.K., S.T.D.M.), Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Groupe Hospitalier Sorbonne Université; Centre de référence pour les maladies vasculaires rares du cerveau et de l'œil (CERVCO) and Centre Neurovascular Translationnel (CNVT) (D.H., A.J., S.R., C.M., S.G., A.T., F.F., H.C.), AP-HP, Paris; and INSERM U1141 - FHU NeuroVasc (D.H., S.G., H.C.), Université Paris Cité, France
| |
Collapse
|
2
|
Garbarino S, Tur C, Lorenzi M, Pardini M, Piana M, Uccelli A, Arnold DL, Cree BAC, Sormani MP, Bovis F. A data-driven model of disability progression in progressive multiple sclerosis. Brain Commun 2024; 7:fcae434. [PMID: 39777254 PMCID: PMC11704797 DOI: 10.1093/braincomms/fcae434] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 10/28/2024] [Accepted: 12/01/2024] [Indexed: 01/11/2025] Open
Abstract
This study applies the Gaussian process progression model, a Bayesian data-driven disease progression model, to analyse the evolution of primary progressive multiple sclerosis. Utilizing data from 1521 primary progressive multiple sclerosis participants collected within the International Progressive Multiple Sclerosis Alliance Project, the analysis includes 18 581 longitudinal time-points (average follow-up time: 28.2 months) of disability assessments including the expanded disability status scale, symbol digit modalities, timed 25-foot-walk, 9-hole-peg test and of MRI metrics such as T1 and T2 lesion volume and normalized brain volume. From these data, Gaussian process progression model infers a data-driven description of the progression common to all individuals, alongside scores measuring the individual progression rates relative to the population, spanning ∼50 years of disease duration. Along this timeline, Gaussian process progression model identifies an initial steep worsening of the expanded disability status scale that stabilizes after ∼30 years of disease duration, suggesting its diminished utility in monitoring disease progression beyond this time. Conversely, it underscores the slower evolution of normalized brain volume across the disease duration. The individual progression rates estimated by Gaussian process progression model can be used to identify three distinct sub-groups within the primary progressive multiple sclerosis population: a normative group (76% of the population) and two 'outlier' sub-groups displaying either accelerated (13% of the population) or decelerated (11%) progression compared to the normative one. Notably, fast progressors exhibit older age at symptom onset (38.5 versus 35.0, P < 0.0001), a higher prevalence of males (61.1% versus 48.5%, P = 0.013) and a higher lesion volumes both in T1 (4.1 versus 0.6, P < 0.0001) and T2 (16.5 versus 7.9, P < 0.0001) compared to slow progressors. Prognostically, fast progressors demonstrate a significantly worse prognosis, with double the risk of experiencing a 3-month confirmed disease progression on expanded disability status scale compared to the normative population according to Cox proportional hazard modelling (HR = 2.09, 95% CI: 1.66-2.62, P < 0.0001) and a shorter median time from the onset of disease symptoms to reaching a confirmed expanded disability status scale 6 (95% CI: 5.83-7.68 years, P < 0.0001). External validation on a test set comprising 227 primary progressive multiple sclerosis participants from the SPI2 trial produced consistent results, with slow progressors exhibiting a reduced risk of experiencing 3-month confirmed disease progression determined through expanded disability status scale (HR = 0.21), while fast progressors facing an increased risk (HR = 1.45). This study contributes to our understanding of disability accrual in primary progressive multiple sclerosis, integrating diverse disability assessments and MRI measurements. Moreover, the identification of distinct sub-groups underscores the heterogeneity in progression rates among patients, offering invaluable insights for patient stratification and monitoring in clinical trials, potentially facilitating more targeted and personalized interventions.
Collapse
Affiliation(s)
- Sara Garbarino
- Life Science Computational laboratory, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
- MIDA, Dipartimento di Matematica, Università di Genova, 16146 Genoa, Italy
| | - Carmen Tur
- Multiple Sclerosis Centre of Catalonia (Cemcat), Department of Neurology, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Marco Lorenzi
- Universitè Côte d’Azur, Inria, Epione Research Project, 06902 Sophia Antipolis, France
| | - Matteo Pardini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Università di Genova, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Michele Piana
- Life Science Computational laboratory, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
- MIDA, Dipartimento di Matematica, Università di Genova, 16146 Genoa, Italy
| | - Antonio Uccelli
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Università di Genova, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | | | - Bruce A C Cree
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, 94143 San Francisco, USA
| | - Maria Pia Sormani
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Department of Health Sciences (DISSAL), Università di Genova, 16132 Genoa, Italy
| | - Francesca Bovis
- Department of Health Sciences (DISSAL), Università di Genova, 16132 Genoa, Italy
| |
Collapse
|
3
|
Zhang C, An L, Wulan N, Nguyen KN, Orban C, Chen P, Chen C, Zhou JH, Liu K, Yeo BT. Cross-dataset Evaluation of Dementia Longitudinal Progression Prediction Models. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.18.24317513. [PMID: 39606367 PMCID: PMC11601715 DOI: 10.1101/2024.11.18.24317513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Accurate Alzheimer's Disease (AD) progression prediction is essential for early intervention. The TADPOLE challenge, involving 92 algorithms, used multimodal biomarkers to predict future clinical diagnosis, cognition, and ventricular volume. The winning algorithm, FROG, utilized a Longitudinal-to-Cross-sectional (L2C) transformation to convert variable longitudinal histories into fixed-length feature vectors, which contrasted with most existing approaches that fitted models to entire longitudinal histories, e.g., AD Course Map (AD-Map) and minimal recurrent neural networks (MinimalRNN). The TADPOLE challenge only utilized the Alzheimer's Disease Neuroimaging Initiative (ADNI) dataset. To evaluate FROG's generalizability, we trained it on the ADNI dataset and tested it on three external datasets covering 2,312 participants and 13,200 timepoints. We also introduced two FROG variants. One variant, L2C feedforward neural network (L2C-FNN), unified all XGBoost models used by the original FROG with an FNN. Across external datasets, L2C-FNN and AD-Map were the best for predicting cognition and ventricular volume. For clinical diagnosis prediction, L2C-FNN was the best, while AD-Map was the worst. L2C-FNN compared favorably with other approaches regardless of the number of observed timepoints, and when predicting from 0 to 6 years into the future, underscoring its potential for long-term dementia progression prediction. Pretrained ADNI models are publicly available: GITHUB_LINK.
Collapse
Affiliation(s)
- Chen Zhang
- Centre for Sleep and Cognition (CSC) & Centre for Translational Magnetic Resonance Research (TMR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
| | - Lijun An
- Centre for Sleep and Cognition (CSC) & Centre for Translational Magnetic Resonance Research (TMR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
| | - Naren Wulan
- Centre for Sleep and Cognition (CSC) & Centre for Translational Magnetic Resonance Research (TMR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
| | - Kim-Ngan Nguyen
- Centre for Sleep and Cognition (CSC) & Centre for Translational Magnetic Resonance Research (TMR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Csaba Orban
- Centre for Sleep and Cognition (CSC) & Centre for Translational Magnetic Resonance Research (TMR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
| | - Pansheng Chen
- Centre for Sleep and Cognition (CSC) & Centre for Translational Magnetic Resonance Research (TMR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
| | - Christopher Chen
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Juan Helen Zhou
- Centre for Sleep and Cognition (CSC) & Centre for Translational Magnetic Resonance Research (TMR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- Department of Medicine, Healthy Longevity Translational Research Programme, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Integrative Sciences and Engineering Programme (ISEP), National University of Singapore
| | | | - B.T. Thomas Yeo
- Centre for Sleep and Cognition (CSC) & Centre for Translational Magnetic Resonance Research (TMR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- Department of Medicine, Healthy Longevity Translational Research Programme, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
- Integrative Sciences and Engineering Programme (ISEP), National University of Singapore
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | | | | |
Collapse
|
4
|
van der Veere PJ, Hoogland J, Visser LNC, Van Harten AC, Rhodius-Meester HF, Sikkes SAM, Venkatraghavan V, Barkhof F, Teunissen CE, van de Giessen E, Berkhof J, Van Der Flier WM. Predicting Cognitive Decline in Amyloid-Positive Patients With Mild Cognitive Impairment or Mild Dementia. Neurology 2024; 103:e209605. [PMID: 38986053 PMCID: PMC11238942 DOI: 10.1212/wnl.0000000000209605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Cognitive decline rates in Alzheimer disease (AD) vary greatly. Disease-modifying treatments may alter cognitive decline trajectories, rendering their prediction increasingly relevant. We aimed to construct clinically applicable prediction models of cognitive decline in amyloid-positive patients with mild cognitive impairment (MCI) or mild dementia. METHODS From the Amsterdam Dementia Cohort, we selected amyloid-positive participants with MCI or mild dementia and at least 2 longitudinal Mini-Mental State Examination (MMSE) measurements. Amyloid positivity was based on CSF AD biomarker concentrations or amyloid PET. We used linear mixed modeling to predict MMSE over time, describing trajectories using a cubic time curve and interactions between linear time and the baseline predictors age, sex, baseline MMSE, APOE ε4 dose, CSF β-amyloid (Aβ) 1-42 and pTau, and MRI total brain and hippocampal volume. Backward selection was used to reduce model complexity. These models can predict MMSE over follow-up or the time to an MMSE value. MCI and mild dementia were modeled separately. Internal 5-fold cross-validation was performed to calculate the explained variance (R2). RESULTS In total, 961 participants were included (age 65 ± 7 years, 49% female), 310 had MCI (MMSE 26 ± 2) and 651 had mild dementia (MMSE 22 ± 4), with 4 ± 2 measurements over 2 (interquartile range 1-4) years. Cognitive decline rates increased over time for both MCI and mild dementia (model comparisons linear vs squared vs cubic time fit; p < 0.05 favoring a cubic fit). For MCI, backward selection retained age, sex, and CSF Aβ1-42 and pTau concentrations as time-varying effects altering the MMSE trajectory. For mild dementia, retained time-varying effects were Aβ1-42, age, APOE ε4, and baseline MMSE. R2 was 0.15 for the MCI model and 0.26 for mild dementia in internal cross-validation. A hypothetical patient with MCI, baseline MMSE 28, and CSF Aβ1-42 of 925 pg/mL was predicted to reach an MMSE of 20 after 6.0 years (95% CI 5.4-6.7) and after 8.6 years with a hypothetical treatment reducing decline by 30%. DISCUSSION We constructed models for MCI and mild dementia that predict MMSE over time. These models could inform patients about their potential cognitive trajectory and the remaining uncertainty and aid in conversations about individualized potential treatment effects.
Collapse
Affiliation(s)
- Pieter J van der Veere
- From the Alzheimer Center and Department of Neurology (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., S.A.M.S., V.V., W.M.V.D.F.), and Department of Epidemiology and Biostatistics (P.J.v.d.V., J.H., L.N.C.V., J.B., W.M.V.D.F.), Amsterdam Neuroscience, VU University Medical Center; Amsterdam Neuroscience (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., V.V., C.E.T., E.G., W.M.V.D.F.), Neurodegeneration the Netherlands; Division of Clinical Geriatrics (L.N.C.V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Medical Psychology (L.N.C.V.), Amsterdam UMC Location AMC, University of Amsterdam; Amsterdam Public Health (L.N.C.V.), Quality of Care, Personalized Medicine; Internal Medicine (H.F.R.-M.), Geriatric Medicine Section, Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC Location VUmc; Department of Clinical, Neuro and Developmental Psychology (S.A.M.S.), Faculty of Movement and Behavioral Sciences, VU University; Department of Radiology & Nuclear Medicine (F.B., E.G.), Amsterdam UMC, Vrije Universiteit, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; and Neurochemistry Laboratory and Biobank (C.E.T.), Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, the Netherlands
| | - Jeroen Hoogland
- From the Alzheimer Center and Department of Neurology (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., S.A.M.S., V.V., W.M.V.D.F.), and Department of Epidemiology and Biostatistics (P.J.v.d.V., J.H., L.N.C.V., J.B., W.M.V.D.F.), Amsterdam Neuroscience, VU University Medical Center; Amsterdam Neuroscience (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., V.V., C.E.T., E.G., W.M.V.D.F.), Neurodegeneration the Netherlands; Division of Clinical Geriatrics (L.N.C.V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Medical Psychology (L.N.C.V.), Amsterdam UMC Location AMC, University of Amsterdam; Amsterdam Public Health (L.N.C.V.), Quality of Care, Personalized Medicine; Internal Medicine (H.F.R.-M.), Geriatric Medicine Section, Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC Location VUmc; Department of Clinical, Neuro and Developmental Psychology (S.A.M.S.), Faculty of Movement and Behavioral Sciences, VU University; Department of Radiology & Nuclear Medicine (F.B., E.G.), Amsterdam UMC, Vrije Universiteit, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; and Neurochemistry Laboratory and Biobank (C.E.T.), Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, the Netherlands
| | - Leonie N C Visser
- From the Alzheimer Center and Department of Neurology (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., S.A.M.S., V.V., W.M.V.D.F.), and Department of Epidemiology and Biostatistics (P.J.v.d.V., J.H., L.N.C.V., J.B., W.M.V.D.F.), Amsterdam Neuroscience, VU University Medical Center; Amsterdam Neuroscience (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., V.V., C.E.T., E.G., W.M.V.D.F.), Neurodegeneration the Netherlands; Division of Clinical Geriatrics (L.N.C.V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Medical Psychology (L.N.C.V.), Amsterdam UMC Location AMC, University of Amsterdam; Amsterdam Public Health (L.N.C.V.), Quality of Care, Personalized Medicine; Internal Medicine (H.F.R.-M.), Geriatric Medicine Section, Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC Location VUmc; Department of Clinical, Neuro and Developmental Psychology (S.A.M.S.), Faculty of Movement and Behavioral Sciences, VU University; Department of Radiology & Nuclear Medicine (F.B., E.G.), Amsterdam UMC, Vrije Universiteit, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; and Neurochemistry Laboratory and Biobank (C.E.T.), Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, the Netherlands
| | - Argonde C Van Harten
- From the Alzheimer Center and Department of Neurology (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., S.A.M.S., V.V., W.M.V.D.F.), and Department of Epidemiology and Biostatistics (P.J.v.d.V., J.H., L.N.C.V., J.B., W.M.V.D.F.), Amsterdam Neuroscience, VU University Medical Center; Amsterdam Neuroscience (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., V.V., C.E.T., E.G., W.M.V.D.F.), Neurodegeneration the Netherlands; Division of Clinical Geriatrics (L.N.C.V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Medical Psychology (L.N.C.V.), Amsterdam UMC Location AMC, University of Amsterdam; Amsterdam Public Health (L.N.C.V.), Quality of Care, Personalized Medicine; Internal Medicine (H.F.R.-M.), Geriatric Medicine Section, Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC Location VUmc; Department of Clinical, Neuro and Developmental Psychology (S.A.M.S.), Faculty of Movement and Behavioral Sciences, VU University; Department of Radiology & Nuclear Medicine (F.B., E.G.), Amsterdam UMC, Vrije Universiteit, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; and Neurochemistry Laboratory and Biobank (C.E.T.), Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, the Netherlands
| | - Hanneke F Rhodius-Meester
- From the Alzheimer Center and Department of Neurology (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., S.A.M.S., V.V., W.M.V.D.F.), and Department of Epidemiology and Biostatistics (P.J.v.d.V., J.H., L.N.C.V., J.B., W.M.V.D.F.), Amsterdam Neuroscience, VU University Medical Center; Amsterdam Neuroscience (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., V.V., C.E.T., E.G., W.M.V.D.F.), Neurodegeneration the Netherlands; Division of Clinical Geriatrics (L.N.C.V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Medical Psychology (L.N.C.V.), Amsterdam UMC Location AMC, University of Amsterdam; Amsterdam Public Health (L.N.C.V.), Quality of Care, Personalized Medicine; Internal Medicine (H.F.R.-M.), Geriatric Medicine Section, Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC Location VUmc; Department of Clinical, Neuro and Developmental Psychology (S.A.M.S.), Faculty of Movement and Behavioral Sciences, VU University; Department of Radiology & Nuclear Medicine (F.B., E.G.), Amsterdam UMC, Vrije Universiteit, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; and Neurochemistry Laboratory and Biobank (C.E.T.), Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, the Netherlands
| | - Sietske A M Sikkes
- From the Alzheimer Center and Department of Neurology (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., S.A.M.S., V.V., W.M.V.D.F.), and Department of Epidemiology and Biostatistics (P.J.v.d.V., J.H., L.N.C.V., J.B., W.M.V.D.F.), Amsterdam Neuroscience, VU University Medical Center; Amsterdam Neuroscience (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., V.V., C.E.T., E.G., W.M.V.D.F.), Neurodegeneration the Netherlands; Division of Clinical Geriatrics (L.N.C.V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Medical Psychology (L.N.C.V.), Amsterdam UMC Location AMC, University of Amsterdam; Amsterdam Public Health (L.N.C.V.), Quality of Care, Personalized Medicine; Internal Medicine (H.F.R.-M.), Geriatric Medicine Section, Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC Location VUmc; Department of Clinical, Neuro and Developmental Psychology (S.A.M.S.), Faculty of Movement and Behavioral Sciences, VU University; Department of Radiology & Nuclear Medicine (F.B., E.G.), Amsterdam UMC, Vrije Universiteit, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; and Neurochemistry Laboratory and Biobank (C.E.T.), Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, the Netherlands
| | - Vikram Venkatraghavan
- From the Alzheimer Center and Department of Neurology (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., S.A.M.S., V.V., W.M.V.D.F.), and Department of Epidemiology and Biostatistics (P.J.v.d.V., J.H., L.N.C.V., J.B., W.M.V.D.F.), Amsterdam Neuroscience, VU University Medical Center; Amsterdam Neuroscience (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., V.V., C.E.T., E.G., W.M.V.D.F.), Neurodegeneration the Netherlands; Division of Clinical Geriatrics (L.N.C.V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Medical Psychology (L.N.C.V.), Amsterdam UMC Location AMC, University of Amsterdam; Amsterdam Public Health (L.N.C.V.), Quality of Care, Personalized Medicine; Internal Medicine (H.F.R.-M.), Geriatric Medicine Section, Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC Location VUmc; Department of Clinical, Neuro and Developmental Psychology (S.A.M.S.), Faculty of Movement and Behavioral Sciences, VU University; Department of Radiology & Nuclear Medicine (F.B., E.G.), Amsterdam UMC, Vrije Universiteit, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; and Neurochemistry Laboratory and Biobank (C.E.T.), Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, the Netherlands
| | - Frederik Barkhof
- From the Alzheimer Center and Department of Neurology (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., S.A.M.S., V.V., W.M.V.D.F.), and Department of Epidemiology and Biostatistics (P.J.v.d.V., J.H., L.N.C.V., J.B., W.M.V.D.F.), Amsterdam Neuroscience, VU University Medical Center; Amsterdam Neuroscience (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., V.V., C.E.T., E.G., W.M.V.D.F.), Neurodegeneration the Netherlands; Division of Clinical Geriatrics (L.N.C.V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Medical Psychology (L.N.C.V.), Amsterdam UMC Location AMC, University of Amsterdam; Amsterdam Public Health (L.N.C.V.), Quality of Care, Personalized Medicine; Internal Medicine (H.F.R.-M.), Geriatric Medicine Section, Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC Location VUmc; Department of Clinical, Neuro and Developmental Psychology (S.A.M.S.), Faculty of Movement and Behavioral Sciences, VU University; Department of Radiology & Nuclear Medicine (F.B., E.G.), Amsterdam UMC, Vrije Universiteit, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; and Neurochemistry Laboratory and Biobank (C.E.T.), Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, the Netherlands
| | - Charlotte E Teunissen
- From the Alzheimer Center and Department of Neurology (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., S.A.M.S., V.V., W.M.V.D.F.), and Department of Epidemiology and Biostatistics (P.J.v.d.V., J.H., L.N.C.V., J.B., W.M.V.D.F.), Amsterdam Neuroscience, VU University Medical Center; Amsterdam Neuroscience (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., V.V., C.E.T., E.G., W.M.V.D.F.), Neurodegeneration the Netherlands; Division of Clinical Geriatrics (L.N.C.V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Medical Psychology (L.N.C.V.), Amsterdam UMC Location AMC, University of Amsterdam; Amsterdam Public Health (L.N.C.V.), Quality of Care, Personalized Medicine; Internal Medicine (H.F.R.-M.), Geriatric Medicine Section, Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC Location VUmc; Department of Clinical, Neuro and Developmental Psychology (S.A.M.S.), Faculty of Movement and Behavioral Sciences, VU University; Department of Radiology & Nuclear Medicine (F.B., E.G.), Amsterdam UMC, Vrije Universiteit, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; and Neurochemistry Laboratory and Biobank (C.E.T.), Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, the Netherlands
| | - Elsmarieke van de Giessen
- From the Alzheimer Center and Department of Neurology (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., S.A.M.S., V.V., W.M.V.D.F.), and Department of Epidemiology and Biostatistics (P.J.v.d.V., J.H., L.N.C.V., J.B., W.M.V.D.F.), Amsterdam Neuroscience, VU University Medical Center; Amsterdam Neuroscience (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., V.V., C.E.T., E.G., W.M.V.D.F.), Neurodegeneration the Netherlands; Division of Clinical Geriatrics (L.N.C.V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Medical Psychology (L.N.C.V.), Amsterdam UMC Location AMC, University of Amsterdam; Amsterdam Public Health (L.N.C.V.), Quality of Care, Personalized Medicine; Internal Medicine (H.F.R.-M.), Geriatric Medicine Section, Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC Location VUmc; Department of Clinical, Neuro and Developmental Psychology (S.A.M.S.), Faculty of Movement and Behavioral Sciences, VU University; Department of Radiology & Nuclear Medicine (F.B., E.G.), Amsterdam UMC, Vrije Universiteit, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; and Neurochemistry Laboratory and Biobank (C.E.T.), Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, the Netherlands
| | - Johannes Berkhof
- From the Alzheimer Center and Department of Neurology (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., S.A.M.S., V.V., W.M.V.D.F.), and Department of Epidemiology and Biostatistics (P.J.v.d.V., J.H., L.N.C.V., J.B., W.M.V.D.F.), Amsterdam Neuroscience, VU University Medical Center; Amsterdam Neuroscience (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., V.V., C.E.T., E.G., W.M.V.D.F.), Neurodegeneration the Netherlands; Division of Clinical Geriatrics (L.N.C.V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Medical Psychology (L.N.C.V.), Amsterdam UMC Location AMC, University of Amsterdam; Amsterdam Public Health (L.N.C.V.), Quality of Care, Personalized Medicine; Internal Medicine (H.F.R.-M.), Geriatric Medicine Section, Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC Location VUmc; Department of Clinical, Neuro and Developmental Psychology (S.A.M.S.), Faculty of Movement and Behavioral Sciences, VU University; Department of Radiology & Nuclear Medicine (F.B., E.G.), Amsterdam UMC, Vrije Universiteit, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; and Neurochemistry Laboratory and Biobank (C.E.T.), Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, the Netherlands
| | - Wiesje M Van Der Flier
- From the Alzheimer Center and Department of Neurology (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., S.A.M.S., V.V., W.M.V.D.F.), and Department of Epidemiology and Biostatistics (P.J.v.d.V., J.H., L.N.C.V., J.B., W.M.V.D.F.), Amsterdam Neuroscience, VU University Medical Center; Amsterdam Neuroscience (P.J.v.d.V., L.N.C.V., A.C.V.H., H.F.R.-M., V.V., C.E.T., E.G., W.M.V.D.F.), Neurodegeneration the Netherlands; Division of Clinical Geriatrics (L.N.C.V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Medical Psychology (L.N.C.V.), Amsterdam UMC Location AMC, University of Amsterdam; Amsterdam Public Health (L.N.C.V.), Quality of Care, Personalized Medicine; Internal Medicine (H.F.R.-M.), Geriatric Medicine Section, Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC Location VUmc; Department of Clinical, Neuro and Developmental Psychology (S.A.M.S.), Faculty of Movement and Behavioral Sciences, VU University; Department of Radiology & Nuclear Medicine (F.B., E.G.), Amsterdam UMC, Vrije Universiteit, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; and Neurochemistry Laboratory and Biobank (C.E.T.), Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, the Netherlands
| |
Collapse
|
5
|
Benani A, Vibert J, Demuth S. [Synthetic data in medicine: Generation, evaluation and limits]. Med Sci (Paris) 2024; 40:661-664. [PMID: 39303119 DOI: 10.1051/medsci/2024091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
Abstract
Recent technological advances in data science hold great promise in medicine. Large-sized high-quality datasets are essential but often difficult to obtain due to privacy, cost, and practical challenges. Here, we discuss synthetic data's generation, evaluation, and regulation, highlighting its current applications and limits.
Collapse
Affiliation(s)
- Alaedine Benani
- Service de méde cine vasculaire, hôpital européen Georges Pompidou (HEGP), AP-HP, Université Paris-Cité, Paris, France - Zoī, Paris, France
| | - Julien Vibert
- Département d'innovations thérapeutiques et essais précoces (DITEP), Inserm U981, Gustave Roussy, Villejuif, Paris, France
| | - Stanislas Demuth
- Inserm U1064, CR2TI - Centre de recherche en transplantation et immunologie translationnelle, Nantes Université, Nantes, France - Inserm CIC 1434, Centre d'investigation clinique, Centre hospitalier de Strasbourg, France
| |
Collapse
|
6
|
Yoshioka H, Jin R, Hisaka A, Suzuki H. Disease progression modeling with temporal realignment: An emerging approach to deepen knowledge on chronic diseases. Pharmacol Ther 2024; 259:108655. [PMID: 38710372 DOI: 10.1016/j.pharmthera.2024.108655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/22/2024] [Accepted: 05/01/2024] [Indexed: 05/08/2024]
Abstract
The recent development of the first disease-modifying drug for Alzheimer's disease represents a major advancement in dementia treatment. Behind this breakthrough is a quarter century of research efforts to understand the disease not by a particular symptom at a given moment, but by long-term sequential changes in multiple biomarkers. Disease progression modeling with temporal realignment (DPM-TR) is an emerging computational approach proposed with this biomarker-based disease concept. By integrating short-term clinical observations of multiple disease biomarkers in a data-driven manner, DPM-TR provides a way to understand the progression of chronic diseases over decades and predict individual disease stages more accurately. DPM-TR has been developed primarily in the area of neurodegenerative diseases but has recently been extended to non-neurodegenerative diseases, including chronic obstructive pulmonary, autoimmune, and ophthalmologic diseases. This review focuses on opportunities for DPM-TR in clinical practice and drug development and discusses its current status and challenges.
Collapse
Affiliation(s)
- Hideki Yoshioka
- Office of Regulatory Science Research, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan; Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Ryota Jin
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Akihiro Hisaka
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan.
| | - Hiroshi Suzuki
- Executive Director, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan; Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
7
|
Rajagopal SK, Beltz AM, Hampstead BM, Polk TA. Estimating individual trajectories of structural and cognitive decline in mild cognitive impairment for early prediction of progression to dementia of the Alzheimer's type. Sci Rep 2024; 14:12906. [PMID: 38839800 PMCID: PMC11153588 DOI: 10.1038/s41598-024-63301-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 05/27/2024] [Indexed: 06/07/2024] Open
Abstract
Only a third of individuals with mild cognitive impairment (MCI) progress to dementia of the Alzheimer's type (DAT). Identifying biomarkers that distinguish individuals with MCI who will progress to DAT (MCI-Converters) from those who will not (MCI-Non-Converters) remains a key challenge in the field. In our study, we evaluate whether the individual rates of loss of volumes of the Hippocampus and entorhinal cortex (EC) with age in the MCI stage can predict progression to DAT. Using data from 758 MCI patients in the Alzheimer's Disease Neuroimaging Database, we employ Linear Mixed Effects (LME) models to estimate individual trajectories of regional brain volume loss over 12 years on average. Our approach involves three key analyses: (1) mapping age-related volume loss trajectories in MCI-Converters and Non-Converters, (2) using logistic regression to predict progression to DAT based on individual rates of hippocampal and EC volume loss, and (3) examining the relationship between individual estimates of these volumetric changes and cognitive decline across different cognitive functions-episodic memory, visuospatial processing, and executive function. We find that the loss of Hippocampal volume is significantly more rapid in MCI-Converters than Non-Converters, but find no such difference in EC volumes. We also find that the rate of hippocampal volume loss in the MCI stage is a significant predictor of conversion to DAT, while the rate of volume loss in the EC and other additional regions is not. Finally, individual estimates of rates of regional volume loss in both the Hippocampus and EC, and other additional regions, correlate strongly with individual rates of cognitive decline. Across all analyses, we find significant individual variation in the initial volumes and the rates of changes in volume with age in individuals with MCI. This study highlights the importance of personalized approaches in predicting AD progression, offering insights for future research and intervention strategies.
Collapse
Affiliation(s)
| | - Adriene M Beltz
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - Benjamin M Hampstead
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
- VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Thad A Polk
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Kumar V, Jangid K, Kumar N, Kumar V, Kumar V. 3D-QSAR-based pharmacophore modelling of quinazoline derivatives for the identification of acetylcholinesterase inhibitors through virtual screening, molecular docking, molecular dynamics and DFT studies. J Biomol Struct Dyn 2024:1-15. [PMID: 38329085 DOI: 10.1080/07391102.2024.2313157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/12/2023] [Indexed: 02/09/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurological disorder responsible for the cognitive dysfunction and cognitive impairment in the patients. Acetylcholinesterase inhibitors (AChEIs) are used to treat AD however, these only provided symptomatic relief and more efficient drug molecules are desired for the effective treatment of the disease. In this article, ligand-based drug-designing strategy was used to develop and validate a field-based 3D-QSAR pharmacophore model on quinazoline-based AChEIs reported in the literature. The validated pharmacophore model (AAAHR_1) was used as a prefilter to screen an ASINEX database via virtual screening workflow (VSW). The hits generated were subjected to MM-GBSA to identify potential AChEIs and top three scoring molecules (BAS 05264565, LEG 12727144 and SYN 22339886) were evaluated for thermodynamic stability at the target site using molecular dynamic simulations. Additionally, DFT study was performed to predict the reactivity of lead molecules towards acetylcholinesterase (AChE). Thus, by utilising various computational tools, three molecules were identified as potent AChEIs that can be developed as potential drug candidates for the treatment of AD.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Chemistry, Laboratory of Organic and Medicinal Chemistry, Central University of Punjab, Bathinda, India
| | - Kailash Jangid
- Department of Chemistry, Laboratory of Organic and Medicinal Chemistry, Central University of Punjab, Bathinda, India
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, India
| | - Naveen Kumar
- Department of Chemistry, Laboratory of Organic and Medicinal Chemistry, Central University of Punjab, Bathinda, India
| | - Vinay Kumar
- Department of Chemistry, Laboratory of Organic and Medicinal Chemistry, Central University of Punjab, Bathinda, India
| | - Vinod Kumar
- Department of Chemistry, Laboratory of Organic and Medicinal Chemistry, Central University of Punjab, Bathinda, India
| |
Collapse
|
9
|
Young AL, Oxtoby NP, Garbarino S, Fox NC, Barkhof F, Schott JM, Alexander DC. Data-driven modelling of neurodegenerative disease progression: thinking outside the black box. Nat Rev Neurosci 2024; 25:111-130. [PMID: 38191721 DOI: 10.1038/s41583-023-00779-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2023] [Indexed: 01/10/2024]
Abstract
Data-driven disease progression models are an emerging set of computational tools that reconstruct disease timelines for long-term chronic diseases, providing unique insights into disease processes and their underlying mechanisms. Such methods combine a priori human knowledge and assumptions with large-scale data processing and parameter estimation to infer long-term disease trajectories from short-term data. In contrast to 'black box' machine learning tools, data-driven disease progression models typically require fewer data and are inherently interpretable, thereby aiding disease understanding in addition to enabling classification, prediction and stratification. In this Review, we place the current landscape of data-driven disease progression models in a general framework and discuss their enhanced utility for constructing a disease timeline compared with wider machine learning tools that construct static disease profiles. We review the insights they have enabled across multiple neurodegenerative diseases, notably Alzheimer disease, for applications such as determining temporal trajectories of disease biomarkers, testing hypotheses about disease mechanisms and uncovering disease subtypes. We outline key areas for technological development and translation to a broader range of neuroscience and non-neuroscience applications. Finally, we discuss potential pathways and barriers to integrating disease progression models into clinical practice and trial settings.
Collapse
Affiliation(s)
- Alexandra L Young
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK.
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Neil P Oxtoby
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK.
| | - Sara Garbarino
- Life Science Computational Laboratory, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Nick C Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Frederik Barkhof
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Jonathan M Schott
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Daniel C Alexander
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| |
Collapse
|
10
|
Lee EY, Kim J, Prado-Rico JM, Du G, Lewis MM, Kong L, Yanosky JD, Eslinger P, Kim BG, Hong YS, Mailman RB, Huang X. Effects of mixed metal exposures on MRI diffusion features in the medial temporal lobe. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.07.18.23292828. [PMID: 37503124 PMCID: PMC10371112 DOI: 10.1101/2023.07.18.23292828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Background Environmental exposure to metal mixtures is common and may be associated with increased risk for neurodegenerative disorders including Alzheimer's disease. Objective This study examined associations of mixed metal exposures with medial temporal lobe (MTL) MRI structural metrics and neuropsychological performance. Methods Metal exposure history, whole blood metal, and neuropsychological tests were obtained from subjects with/without a history of mixed metal exposure from welding fumes (42 exposed subjects; 31 controls). MTL structures (hippocampus, entorhinal and parahippocampal cortices) were assessed by morphologic (volume, cortical thickness) and diffusion tensor imaging [mean (MD), axial (AD), radial diffusivity (RD), and fractional anisotropy (FA)] metrics. In exposed subjects, correlation, multiple linear, Bayesian kernel machine regression, and mediation analyses were employed to examine effects of single- or mixed-metal predictor(s) and their interactions on MTL structural and neuropsychological metrics; and on the path from metal exposure to neuropsychological consequences. Results Compared to controls, exposed subjects had higher blood Cu, Fe, K, Mn, Pb, Se, and Zn levels (p's<0.026) and poorer performance in processing/psychomotor speed, executive, and visuospatial domains (p's<0.046). Exposed subjects displayed higher MD, AD, and RD in all MTL ROIs (p's<0.040) and lower FA in entorhinal and parahippocampal cortices (p's<0.033), but not morphological differences. Long-term mixed-metal exposure history indirectly predicted lower processing speed performance via lower parahippocampal FA (p=0.023). Higher whole blood Mn and Cu predicted higher entorhinal diffusivity (p's<0.043) and lower Delayed Story Recall performance (p=0.007) without overall metal mixture or interaction effects. Discussion Mixed metal exposure predicted MTL structural and neuropsychological features that are similar to Alzheimer's disease at-risk populations. These data warrant follow-up as they may illuminate the path for environmental exposure to Alzheimer's disease-related health outcomes.
Collapse
Affiliation(s)
- Eun-Young Lee
- Department of Health Care and Science, Dong-A University, Busan, South-Korea
| | - Juhee Kim
- Department of Health Care and Science, Dong-A University, Busan, South-Korea
| | - Janina Manzieri Prado-Rico
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Guangwei Du
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Mechelle M. Lewis
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
- Department of Pharmacology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Lan Kong
- Department of Public Health Sciences, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Jeff D. Yanosky
- Department of Public Health Sciences, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Paul Eslinger
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Byoung-Gwon Kim
- Department of Preventive Medicine, College of Medicine, Dong-A University, Busan, South Korea
| | - Young-Seoub Hong
- Department of Preventive Medicine, College of Medicine, Dong-A University, Busan, South Korea
| | - Richard B. Mailman
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
- Department of Pharmacology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Xuemei Huang
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
- Department of Pharmacology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
- Department of Radiology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
- Department of Neurosurgery, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
- Department of Kinesiology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| |
Collapse
|
11
|
Di Folco C, Couronné R, Arnulf I, Mangone G, Leu-Semenescu S, Dodet P, Vidailhet M, Corvol JC, Lehéricy S, Durrleman S. Charting Disease Trajectories from Isolated REM Sleep Behavior Disorder to Parkinson's Disease. Mov Disord 2024; 39:64-75. [PMID: 38006282 DOI: 10.1002/mds.29662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/03/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Clinical presentation and progression dynamics are variable in patients with Parkinson's disease (PD). Disease course mapping is an innovative disease modelling technique that summarizes the range of possible disease trajectories and estimates dimensions related to onset, sequence, and speed of progression of disease markers. OBJECTIVE To propose a disease course map for PD and investigate progression profiles in patients with or without rapid eye movement sleep behavioral disorders (RBD). METHODS Data of 919 PD patients and 88 isolated RBD patients from three independent longitudinal cohorts were analyzed (follow-up duration = 5.1; 95% confidence interval, 1.1-8.1] years). Disease course map was estimated by using eight clinical markers (motor and non-motor symptoms) and four imaging markers (dopaminergic denervation). RESULTS PD course map showed that the first changes occurred in the contralateral putamen 13 years before diagnosis, followed by changes in motor symptoms, dysautonomia, sleep-all before diagnosis-and finally cognitive decline at the time of diagnosis. The model showed earlier disease onset, earlier non-motor and later motor symptoms, more rapid progression of cognitive decline in PD patients with RBD than PD patients without RBD. This pattern was even more pronounced in patients with isolated RBD with early changes in sleep, followed by cognition and non-motor symptoms and later changes in motor symptoms. CONCLUSIONS Our findings are consistent with the presence of distinct patterns of progression between patients with and without RBD. Understanding heterogeneity of PD progression is key to decipher the underlying pathophysiology and select homogeneous subgroups of patients for precision medicine. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Cécile Di Folco
- Inria, Centre de Paris, Paris, France
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Raphaël Couronné
- Inria, Centre de Paris, Paris, France
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Isabelle Arnulf
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Graziella Mangone
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Smaranda Leu-Semenescu
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Pauline Dodet
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Marie Vidailhet
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Jean-Christophe Corvol
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Stéphane Lehéricy
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Stanley Durrleman
- Inria, Centre de Paris, Paris, France
- Paris Brain Institute-ICM, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Sorbonne Université, Paris, France
- AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| |
Collapse
|
12
|
Platero C, Tohka J, Strange B. Estimating Dementia Onset: AT(N) Profiles and Predictive Modeling in Mild Cognitive Impairment Patients. Curr Alzheimer Res 2024; 20:778-790. [PMID: 38425106 DOI: 10.2174/0115672050295317240223162312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/06/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Mild Cognitive Impairment (MCI) usually precedes the symptomatic phase of dementia and constitutes a window of opportunities for preventive therapies. OBJECTIVES The objective of this study was to predict the time an MCI patient has left to reach dementia and obtain the most likely natural history in the progression of MCI towards dementia. METHODS This study was conducted on 633 MCI patients and 145 subjects with dementia through 4726 visits over 15 years from Alzheimer Disease Neuroimaging Initiative (ADNI) cohort. A combination of data from AT(N) profiles at baseline and longitudinal predictive modeling was applied. A data-driven approach was proposed for categorical diagnosis prediction and timeline estimation of cognitive decline progression, which combined supervised and unsupervised learning techniques. RESULTS A reduced vector of only neuropsychological measures was selected for training the models. At baseline, this approach had high performance in detecting subjects at high risk of converting from MCI to dementia in the coming years. Furthermore, a Disease Progression Model (DPM) was built and also verified using three metrics. As a result of the DPM focused on the studied population, it was inferred that amyloid pathology (A+) appears about 7 years before dementia, and tau pathology (T+) and neurodegeneration (N+) occur almost simultaneously, between 3 and 4 years before dementia. In addition, MCI-A+ subjects were shown to progress more rapidly to dementia compared to MCI-A- subjects. CONCLUSION Based on proposed natural histories and cross-sectional and longitudinal analysis of AD markers, the results indicated that only a single cerebrospinal fluid sample is necessary during the prodromal phase of AD. Prediction from MCI into dementia and its timeline can be achieved exclusively through neuropsychological measures.
Collapse
Affiliation(s)
- Carlos Platero
- Health Science Technology Group, Technical University of Madrid, 28012 Madrid, Spain
| | - Jussi Tohka
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Bryan Strange
- Laboratory for Clinical Neuroscience, CTB, Technical University of Madrid, IdISSC, Madrid, Spain
- Alzheimer Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center, Madrid, Spain
| |
Collapse
|
13
|
Veitch DP, Weiner MW, Miller M, Aisen PS, Ashford MA, Beckett LA, Green RC, Harvey D, Jack CR, Jagust W, Landau SM, Morris JC, Nho KT, Nosheny R, Okonkwo O, Perrin RJ, Petersen RC, Rivera Mindt M, Saykin A, Shaw LM, Toga AW, Tosun D. The Alzheimer's Disease Neuroimaging Initiative in the era of Alzheimer's disease treatment: A review of ADNI studies from 2021 to 2022. Alzheimers Dement 2024; 20:652-694. [PMID: 37698424 PMCID: PMC10841343 DOI: 10.1002/alz.13449] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 09/13/2023]
Abstract
The Alzheimer's Disease Neuroimaging Initiative (ADNI) aims to improve Alzheimer's disease (AD) clinical trials. Since 2006, ADNI has shared clinical, neuroimaging, and cognitive data, and biofluid samples. We used conventional search methods to identify 1459 publications from 2021 to 2022 using ADNI data/samples and reviewed 291 impactful studies. This review details how ADNI studies improved disease progression understanding and clinical trial efficiency. Advances in subject selection, detection of treatment effects, harmonization, and modeling improved clinical trials and plasma biomarkers like phosphorylated tau showed promise for clinical use. Biomarkers of amyloid beta, tau, neurodegeneration, inflammation, and others were prognostic with individualized prediction algorithms available online. Studies supported the amyloid cascade, emphasized the importance of neuroinflammation, and detailed widespread heterogeneity in disease, linked to genetic and vascular risk, co-pathologies, sex, and resilience. Biological subtypes were consistently observed. Generalizability of ADNI results is limited by lack of cohort diversity, an issue ADNI-4 aims to address by enrolling a diverse cohort.
Collapse
Affiliation(s)
- Dallas P. Veitch
- Department of Veterans Affairs Medical CenterNorthern California Institute for Research and Education (NCIRE)San FranciscoCaliforniaUSA
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
| | - Michael W. Weiner
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of MedicineUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of Psychiatry and Behavioral SciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Melanie Miller
- Department of Veterans Affairs Medical CenterNorthern California Institute for Research and Education (NCIRE)San FranciscoCaliforniaUSA
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
| | - Paul S. Aisen
- Alzheimer's Therapeutic Research InstituteUniversity of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Miriam A. Ashford
- Department of Veterans Affairs Medical CenterNorthern California Institute for Research and Education (NCIRE)San FranciscoCaliforniaUSA
| | - Laurel A. Beckett
- Division of BiostatisticsDepartment of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | - Robert C. Green
- Division of GeneticsDepartment of MedicineBrigham and Women's HospitalBroad Institute Ariadne Labs and Harvard Medical SchoolBostonMassachusettsUSA
| | - Danielle Harvey
- Division of BiostatisticsDepartment of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | | | - William Jagust
- Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - Susan M. Landau
- Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - John C. Morris
- Knight Alzheimer's Disease Research CenterWashington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
- Department of Pathology and ImmunologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Kwangsik T. Nho
- Department of Radiology and Imaging Sciences and the Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for Computational Biology and BioinformaticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Rachel Nosheny
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of Psychiatry and Behavioral SciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Ozioma Okonkwo
- Wisconsin Alzheimer's Disease Research Center and Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Richard J. Perrin
- Knight Alzheimer's Disease Research CenterWashington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
- Department of Pathology and ImmunologyWashington University School of MedicineSaint LouisMissouriUSA
| | | | - Monica Rivera Mindt
- Department of PsychologyLatin American and Latino Studies InstituteAfrican and African American StudiesFordham UniversityNew YorkNew YorkUSA
- Department of NeurologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Andrew Saykin
- Department of Radiology and Imaging Sciences and the Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine and the PENN Alzheimer's Disease Research CenterCenter for Neurodegenerative ResearchPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Arthur W. Toga
- Laboratory of Neuro ImagingInstitute of Neuroimaging and InformaticsKeck School of Medicine of University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Duygu Tosun
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | | |
Collapse
|
14
|
Ortholand J, Pradat PF, Tezenas du Montcel S, Durrleman S. Interaction of sex and onset site on the disease trajectory of amyotrophic lateral sclerosis. J Neurol 2023; 270:5903-5912. [PMID: 37615751 DOI: 10.1007/s00415-023-11932-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND Studies showed the impact of sex and onset site (spinal or bulbar) on disease onset and survival in ALS. However, they mainly result from cross-sectional or survival analysis, and the interaction of sex and onset site on the different proxies of disease trajectory has not been fully investigated. METHODS We selected all patients with repeated observations in the PRO-ACT database. We divided them into four groups depending on their sex and onset site. We estimated a multivariate disease progression model, named ALS Course Map, to investigate the combined temporal changes of the four sub-scores of the revised ALS functional rating scale (ALSFRSr), the forced vital capacity (FVC), and the body mass index (BMI). We then compared the progression rate, the estimated age at onset, and the relative progression of the outcomes across each group. RESULTS We included 1438 patients from the PRO-ACT database. They were 51% men with spinal onset, 12% men with bulbar onset, 26% women with spinal onset, and 11% women with bulbar onset. We showed a significant influence of both sex and onset site on the ALSFRSr progression. The BMI decreased 8.9 months earlier (95% CI [3.9, 13.8]) in women than men, after correction for the onset site. Among patients with bulbar onset, FVC was impaired 2.6 months earlier (95% CI [0.6, 4.6]) in women. CONCLUSION Using a multivariable disease modelling approach, we showed that sex and onset site are important drivers of the progression of motor function, BMI, and FVC decline.
Collapse
Affiliation(s)
- Juliette Ortholand
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute, ICM, CNRS, InriaInserm, AP-HP, Hôpital de La Pitié Salpêtrière, 75013, Paris, France.
| | - Pierre-François Pradat
- Laboratoire d'Imagerie Biomédicale, Sorbonne Université, CNRS, INSERM, Paris, France
- APHP, Département de Neurologie, Hôpital Pitié-Salpêtrière, Centre Référent SLA, Paris, France
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute Ulster University, C-TRIC, Altnagelvin Hospital, Derry, Londonderry, UK
| | - Sophie Tezenas du Montcel
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute, ICM, CNRS, InriaInserm, AP-HP, Hôpital de La Pitié Salpêtrière, 75013, Paris, France
| | - Stanley Durrleman
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute, ICM, CNRS, InriaInserm, AP-HP, Hôpital de La Pitié Salpêtrière, 75013, Paris, France
| |
Collapse
|
15
|
Lespinasse J, Dufouil C, Proust-Lima C. Disease progression model anchored around clinical diagnosis in longitudinal cohorts: example of Alzheimer's disease and related dementia. BMC Med Res Methodol 2023; 23:199. [PMID: 37670234 PMCID: PMC10478286 DOI: 10.1186/s12874-023-02009-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 08/04/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND Alzheimer's disease and related dementia (ADRD) are characterized by multiple and progressive anatomo-clinical changes including accumulation of abnormal proteins in the brain, brain atrophy and severe cognitive impairment. Understanding the sequence and timing of these changes is of primary importance to gain insight into the disease natural history and ultimately allow earlier diagnosis. Yet, modeling changes over disease course from cohort data is challenging as the usual timescales (time since inclusion, chronological age) are inappropriate and time-to-clinical diagnosis is available on small subsamples of participants with short follow-up durations prior to diagnosis. One solution to circumvent this challenge is to define the disease time as a latent variable. METHODS We developed a multivariate mixed model approach that realigns individual trajectories into the latent disease time to describe disease progression. In contrast with the existing literature, our methodology exploits the clinical diagnosis information as a partially observed and approximate reference to guide the estimation of the latent disease time. The model estimation was carried out in the Bayesian Framework using Stan. We applied the methodology to the MEMENTO study, a French multicentric clinic-based cohort of 2186 participants with 5-year intensive follow-up. Repeated measures of 12 ADRD markers stemmed from cerebrospinal fluid (CSF), brain imaging and cognitive tests were analyzed. RESULTS The estimated latent disease time spanned over twenty years before the clinical diagnosis. Considering the profile of a woman aged 70 with a high level of education and APOE4 carrier (the main genetic risk factor for ADRD), CSF markers of tau proteins accumulation preceded markers of brain atrophy by 5 years and cognitive decline by 10 years. However we observed that individual characteristics could substantially modify the sequence and timing of these changes, in particular for CSF level of A[Formula: see text]. CONCLUSION By leveraging the available clinical diagnosis timing information, our disease progression model does not only realign trajectories into the most homogeneous way. It accounts for the inherent residual inter-individual variability in dementia progression to describe the long-term anatomo-clinical degradations according to the years preceding clinical diagnosis, and to provide clinically meaningful information on the sequence of events. TRIAL REGISTRATION clinicaltrials.gov, NCT01926249. Registered on 16 August 2013.
Collapse
Affiliation(s)
- Jérémie Lespinasse
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, BPH, U1219, 33000, Bordeaux, France
- Inserm, CIC1401-EC, 33000, Bordeaux, France
- Pôle de santé publique, Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000, Bordeaux, France
| | - Carole Dufouil
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, BPH, U1219, 33000, Bordeaux, France
- Inserm, CIC1401-EC, 33000, Bordeaux, France
- Pôle de santé publique, Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000, Bordeaux, France
| | - Cécile Proust-Lima
- Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, BPH, U1219, 33000, Bordeaux, France.
- Inserm, CIC1401-EC, 33000, Bordeaux, France.
| |
Collapse
|
16
|
Saint-Jalmes M, Fedyashov V, Beck D, Baldwin T, Faux NG, Bourgeat P, Fripp J, Masters CL, Goudey B. Disease progression modelling of Alzheimer's disease using probabilistic principal components analysis. Neuroimage 2023; 278:120279. [PMID: 37454702 DOI: 10.1016/j.neuroimage.2023.120279] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/27/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023] Open
Abstract
The recent biological redefinition of Alzheimer's Disease (AD) has spurred the development of statistical models that relate changes in biomarkers with neurodegeneration and worsening condition linked to AD. The ability to measure such changes may facilitate earlier diagnoses for affected individuals and help in monitoring the evolution of their condition. Amongst such statistical tools, disease progression models (DPMs) are quantitative, data-driven methods that specifically attempt to describe the temporal dynamics of biomarkers relevant to AD. Due to the heterogeneous nature of this disease, with patients of similar age experiencing different AD-related changes, a challenge facing longitudinal mixed-effects-based DPMs is the estimation of patient-realigning time-shifts. These time-shifts are indispensable for meaningful biomarker modelling, but may impact fitting time or vary with missing data in jointly estimated models. In this work, we estimate an individual's progression through Alzheimer's disease by combining multiple biomarkers into a single value using a probabilistic formulation of principal components analysis. Our results show that this variable, which summarises AD through observable biomarkers, is remarkably similar to jointly estimated time-shifts when we compute our scores for the baseline visit, on cross-sectional data from the Alzheimer's Disease Neuroimaging Initiative (ADNI). Reproducing the expected properties of clinical datasets, we confirm that estimated scores are robust to missing data or unavailable biomarkers. In addition to cross-sectional insights, we can model the latent variable as an individual progression score by repeating estimations at follow-up examinations and refining long-term estimates as more data is gathered, which would be ideal in a clinical setting. Finally, we verify that our score can be used as a pseudo-temporal scale instead of age to ignore some patient heterogeneity in cohort data and highlight the general trend in expected biomarker evolution in affected individuals.
Collapse
Affiliation(s)
- Martin Saint-Jalmes
- ARC Training Centre in Cognitive Computing for Medical Technologies, University of Melbourne, Carlton, VIC, Australia; The Florey Department of Neuroscience and Mental Health, The University of Melbourne, Australia; The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Australia.
| | - Victor Fedyashov
- ARC Training Centre in Cognitive Computing for Medical Technologies, University of Melbourne, Carlton, VIC, Australia; The Florey Department of Neuroscience and Mental Health, The University of Melbourne, Australia
| | - Daniel Beck
- ARC Training Centre in Cognitive Computing for Medical Technologies, University of Melbourne, Carlton, VIC, Australia; School of Computing and Information Systems, The University of Melbourne, Australia
| | - Timothy Baldwin
- ARC Training Centre in Cognitive Computing for Medical Technologies, University of Melbourne, Carlton, VIC, Australia; School of Computing and Information Systems, The University of Melbourne, Australia; Mohamed bin Zayed University of Artificial Intelligence, Abu Dhabi, United Arab Emirates
| | - Noel G Faux
- ARC Training Centre in Cognitive Computing for Medical Technologies, University of Melbourne, Carlton, VIC, Australia; The Florey Department of Neuroscience and Mental Health, The University of Melbourne, Australia; Melbourne Data Analytics Platform, The University of Melbourne, Australia
| | | | - Jurgen Fripp
- CSIRO Health and Biosecurity, Brisbane, Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Australia
| | - Benjamin Goudey
- ARC Training Centre in Cognitive Computing for Medical Technologies, University of Melbourne, Carlton, VIC, Australia; The Florey Department of Neuroscience and Mental Health, The University of Melbourne, Australia
| |
Collapse
|
17
|
Wijeratne PA, Eshaghi A, Scotton WJ, Kohli M, Aksman L, Oxtoby NP, Pustina D, Warner JH, Paulsen JS, Scahill RI, Sampaio C, Tabrizi SJ, Alexander DC. The temporal event-based model: Learning event timelines in progressive diseases. IMAGING NEUROSCIENCE (CAMBRIDGE, MASS.) 2023; 1:1-19. [PMID: 37719837 PMCID: PMC10503481 DOI: 10.1162/imag_a_00010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 07/15/2023] [Indexed: 09/19/2023]
Abstract
Timelines of events, such as symptom appearance or a change in biomarker value, provide powerful signatures that characterise progressive diseases. Understanding and predicting the timing of events is important for clinical trials targeting individuals early in the disease course when putative treatments are likely to have the strongest effect. However, previous models of disease progression cannot estimate the time between events and provide only an ordering in which they change. Here, we introduce the temporal event-based model (TEBM), a new probabilistic model for inferring timelines of biomarker events from sparse and irregularly sampled datasets. We demonstrate the power of the TEBM in two neurodegenerative conditions: Alzheimer's disease (AD) and Huntington's disease (HD). In both diseases, the TEBM not only recapitulates current understanding of event orderings but also provides unique new ranges of timescales between consecutive events. We reproduce and validate these findings using external datasets in both diseases. We also demonstrate that the TEBM improves over current models; provides unique stratification capabilities; and enriches simulated clinical trials to achieve a power of 80 % with less than half the cohort size compared with random selection. The application of the TEBM naturally extends to a wide range of progressive conditions.
Collapse
Affiliation(s)
- Peter A. Wijeratne
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
- Department of Informatics, University of Sussex, Brighton, United Kingdom
| | - Arman Eshaghi
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, University College London, London, United Kingdom
| | - William J. Scotton
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London, London, United Kingdom
| | - Maitrei Kohli
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| | - Leon Aksman
- Keck School of Medicine, University of Southern California, Los Angeles, California, United States
| | - Neil P. Oxtoby
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| | - Dorian Pustina
- CHDI Management/CHDI Foundation, Princeton, New Jersey, United States
| | - John H. Warner
- CHDI Management/CHDI Foundation, Princeton, New Jersey, United States
| | - Jane S. Paulsen
- Departments of Neurology and Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Rachael I. Scahill
- Huntington’s Disease Centre, Department of Neurodegenerative Disease, University College London, Queen Square, London, United Kingdom
| | - Cristina Sampaio
- CHDI Management/CHDI Foundation, Princeton, New Jersey, United States
| | - Sarah J. Tabrizi
- Huntington’s Disease Centre, Department of Neurodegenerative Disease, University College London, Queen Square, London, United Kingdom
| | - Daniel C. Alexander
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| |
Collapse
|
18
|
Reynolds M, Chaudhary T, Eshaghzadeh Torbati M, Tudorascu DL, Batmanghelich K. ComBat Harmonization: Empirical Bayes versus fully Bayes approaches. Neuroimage Clin 2023; 39:103472. [PMID: 37506457 PMCID: PMC10412957 DOI: 10.1016/j.nicl.2023.103472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023]
Abstract
Studying small effects or subtle neuroanatomical variation requires large-scale sample size data. As a result, combining neuroimaging data from multiple datasets is necessary. Variation in acquisition protocols, magnetic field strength, scanner build, and many other non-biologically related factors can introduce undesirable bias into studies. Hence, harmonization is required to remove the bias-inducing factors from the data. ComBat is one of the most common methods applied to features from structural images. ComBat models the data using a hierarchical Bayesian model and uses the empirical Bayes approach to infer the distribution of the unknown factors. The empirical Bayes harmonization method is computationally efficient and provides valid point estimates. However, it tends to underestimate uncertainty. This paper investigates a new approach, fully Bayesian ComBat, where Monte Carlo sampling is used for statistical inference. When comparing fully Bayesian and empirical Bayesian ComBat, we found Empirical Bayesian ComBat more effectively removed scanner strength information and was much more computationally efficient. Conversely, fully Bayesian ComBat better preserved biological disease and age-related information while performing more accurate harmonization on traveling subjects. The fully Bayesian approach generates a rich posterior distribution, which is useful for generating simulated imaging features for improving classifier performance in a limited data setting. We show the generative capacity of our model for augmenting and improving the detection of patients with Alzheimer's disease. Posterior distributions for harmonized imaging measures can also be used for brain-wide uncertainty comparison and more principled downstream statistical analysis.Code for our new fully Bayesian ComBat extension is available at https://github.com/batmanlab/BayesComBat.
Collapse
Affiliation(s)
- Maxwell Reynolds
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, 5607 Baum Blvd. Suite 500, Pittsburgh, PA 15206, USA.
| | - Tigmanshu Chaudhary
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, 5607 Baum Blvd. Suite 500, Pittsburgh, PA 15206, USA.
| | - Mahbaneh Eshaghzadeh Torbati
- Intelligent System Program, University of Pittsburgh School of Computing and Information, 210 South Bouquet Street, Pittsburgh, PA 15260, USA.
| | - Dana L Tudorascu
- Department of Psychiatry, University of Pittsburgh School of Medicine, 3811 O'Hara Street, Pittsburgh, PA 15213, USA; Department of Biostatistics, University of Pittsburgh, 130 De Soto Street, Pittsburgh, PA 15213, USA.
| | - Kayhan Batmanghelich
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, 5607 Baum Blvd. Suite 500, Pittsburgh, PA 15206, USA.
| |
Collapse
|
19
|
Sauty B, Durrleman S. Impact of sex and APOE- ε4 genotype on patterns of regional brain atrophy in Alzheimer's disease and healthy aging. Front Neurol 2023; 14:1161527. [PMID: 37333001 PMCID: PMC10272760 DOI: 10.3389/fneur.2023.1161527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/19/2023] [Indexed: 06/20/2023] Open
Abstract
Alzheimer's Disease (AD) is a heterogeneous disease that disproportionately affects women and people with the APOE-ε4 susceptibility gene. We aim to describe the not-well-understood influence of both risk factors on the dynamics of brain atrophy in AD and healthy aging. Regional cortical thinning and brain atrophy were modeled over time using non-linear mixed-effect models and the FreeSurfer software with t1-MRI scans from the Alzheimer's Disease Neuroimaging Initiative (N = 1,502 subjects, 6,728 images in total). Covariance analysis was used to disentangle the effect of sex and APOE genotype on the regional onset age and pace of atrophy, while correcting for educational level. A map of the regions mostly affected by neurodegeneration is provided. Results were confirmed on gray matter density data from the SPM software. Women experience faster atrophic rates in the temporal, frontal, parietal lobes and limbic system and earlier onset in the amygdalas, but slightly later onset in the postcentral and cingulate gyri as well as all regions of the basal ganglia and thalamus. APOE-ε4 genotypes leads to earlier and faster atrophy in the temporal, frontal, parietal lobes, and limbic system in AD patients, but not in healthy patients. Higher education was found to slightly delay atrophy in healthy patients, but not for AD patients. A cohort of amyloid positive patients with MCI showed a similar impact of sex as in the healthy cohort, while APOE-ε4 showed similar associations as in the AD cohort. Female sex is as strong a risk factor for AD as APOE-ε4 genotype regarding neurodegeneration. Women experience a sharper atrophy in the later stages of the disease, although not a significantly earlier onset. These findings may have important implications for the development of targeted intervention.
Collapse
|
20
|
Seyedsalehi A, Warrier V, Bethlehem RAI, Perry BI, Burgess S, Murray GK. Educational attainment, structural brain reserve and Alzheimer's disease: a Mendelian randomization analysis. Brain 2023; 146:2059-2074. [PMID: 36310536 PMCID: PMC10151197 DOI: 10.1093/brain/awac392] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 09/01/2022] [Accepted: 09/19/2022] [Indexed: 11/13/2022] Open
Abstract
Higher educational attainment is observationally associated with lower risk of Alzheimer's disease. However, the biological mechanisms underpinning this association remain unclear. The protective effect of education on Alzheimer's disease may be mediated via increased brain reserve. We used two-sample Mendelian randomization to explore putative causal relationships between educational attainment, structural brain reserve as proxied by MRI phenotypes and Alzheimer's disease. Summary statistics were obtained from genome-wide association studies of educational attainment (n = 1 131 881), late-onset Alzheimer's disease (35 274 cases, 59 163 controls) and 15 measures of grey or white matter macro- or micro-structure derived from structural or diffusion MRI (nmax = 33 211). We conducted univariable Mendelian randomization analyses to investigate bidirectional associations between (i) educational attainment and Alzheimer's disease; (ii) educational attainment and imaging-derived phenotypes; and (iii) imaging-derived phenotypes and Alzheimer's disease. Multivariable Mendelian randomization was used to assess whether brain structure phenotypes mediated the effect of education on Alzheimer's disease risk. Genetically proxied educational attainment was inversely associated with Alzheimer's disease (odds ratio per standard deviation increase in genetically predicted years of schooling = 0.70, 95% confidence interval 0.60, 0.80). There were positive associations between genetically predicted educational attainment and four cortical metrics (standard deviation units change in imaging phenotype per one standard deviation increase in genetically predicted years of schooling): surface area 0.30 (95% confidence interval 0.20, 0.40); volume 0.29 (95% confidence interval 0.20, 0.37); intrinsic curvature 0.18 (95% confidence interval 0.11, 0.25); local gyrification index 0.21 (95% confidence interval 0.11, 0.31)]; and inverse associations with cortical intracellular volume fraction [-0.09 (95% confidence interval -0.15, -0.03)] and white matter hyperintensities volume [-0.14 (95% confidence interval -0.23, -0.05)]. Genetically proxied levels of surface area, cortical volume and intrinsic curvature were positively associated with educational attainment [standard deviation units change in years of schooling per one standard deviation increase in respective genetically predicted imaging phenotype: 0.13 (95% confidence interval 0.10, 0.16); 0.15 (95% confidence interval 0.11, 0.19) and 0.12 (95% confidence interval 0.04, 0.19)]. We found no evidence of associations between genetically predicted imaging-derived phenotypes and Alzheimer's disease. The inverse association of genetically predicted educational attainment with Alzheimer's disease did not attenuate after adjusting for imaging-derived phenotypes in multivariable analyses. Our results provide support for a protective causal effect of educational attainment on Alzheimer's disease risk, as well as potential bidirectional causal relationships between education and brain macro- and micro-structure. However, we did not find evidence that these structural markers affect risk of Alzheimer's disease. The protective effect of education on Alzheimer's disease may be mediated via other measures of brain reserve not included in the present study, or by alternative mechanisms.
Collapse
Affiliation(s)
- Aida Seyedsalehi
- Department of Public Health and Primary Care, University of Cambridge, Cambridge CB2 0SR, UK
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford OX3 7JX, UK
| | - Varun Warrier
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge CB2 8AH, UK
| | - Richard A I Bethlehem
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge CB2 8AH, UK
- Brain Mapping Unit, Department of Psychiatry, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Benjamin I Perry
- Department of Psychiatry, University of Cambridge, Cambridge CB2 8AH, UK
- CAMEO, Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge CB4 1PX, UK
| | - Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, Cambridge CB2 0SR, UK
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB2 0BB, UK
| | - Graham K Murray
- Department of Psychiatry, University of Cambridge, Cambridge CB2 8AH, UK
- CAMEO, Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge CB4 1PX, UK
- Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, Australia
| |
Collapse
|
21
|
Chen H, Young A, Oxtoby NP, Barkhof F, Alexander DC, Altmann A. Transferability of Alzheimer's disease progression subtypes to an independent population cohort. Neuroimage 2023; 271:120005. [PMID: 36907283 DOI: 10.1016/j.neuroimage.2023.120005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/22/2023] [Accepted: 03/06/2023] [Indexed: 03/13/2023] Open
Abstract
In the past, methods to subtype or biotype patients using brain imaging data have been developed. However, it is unclear whether and how these trained machine learning models can be successfully applied to population cohorts to study the genetic and lifestyle factors underpinning these subtypes. This work, using the Subtype and Stage Inference (SuStaIn) algorithm, examines the generalisability of data-driven Alzheimer's disease (AD) progression models. We first compared SuStaIn models trained separately on Alzheimer's disease neuroimaging initiative (ADNI) data and an AD-at-risk population constructed from the UK Biobank dataset. We further applied data harmonization techniques to remove cohort effects. Next, we built SuStaIn models on the harmonized datasets, which were then used to subtype and stage subjects in the other harmonized dataset. The first key finding is that three consistent atrophy subtypes were found in both datasets, which match the previously identified subtype progression patterns in AD: 'typical', 'cortical' and 'subcortical'. Next, the subtype agreement was further supported by high consistency in individuals' subtypes and stage assignment based on the different models: more than 92% of the subjects, with reliable subtype assignment in both ADNI and UK Biobank dataset, were assigned to an identical subtype under the model built on the different datasets. The successful transferability of AD atrophy progression subtypes across cohorts capturing different phases of disease development enabled further investigations of associations between AD atrophy subtypes and risk factors. Our study showed that (1) the average age is highest in the typical subtype and lowest in the subcortical subtype; (2) the typical subtype is associated with statistically more-AD-like cerebrospinal fluid biomarkers values in comparison to the other two subtypes; and (3) in comparison to the subcortical subtype, the cortical subtype subjects are more likely to associate with prescription of cholesterol and high blood pressure medications. In summary, we presented cross-cohort consistent recovery of AD atrophy subtypes, showing how the same subtypes arise even in cohorts capturing substantially different disease phases. Our study opened opportunities for future detailed investigations of atrophy subtypes with a broad range of early risk factors, which will potentially lead to a better understanding of the disease aetiology and the role of lifestyle and behaviour on AD.
Collapse
Affiliation(s)
- Hanyi Chen
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering and Department of Computer Science, University College London, UK
| | - Alexandra Young
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Neil P Oxtoby
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering and Department of Computer Science, University College London, UK
| | - Frederik Barkhof
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering and Department of Computer Science, University College London, UK; Queen Square Institute of Neurology, University College London, UK; Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, The Netherlands
| | - Daniel C Alexander
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering and Department of Computer Science, University College London, UK
| | - Andre Altmann
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering and Department of Computer Science, University College London, UK.
| | | |
Collapse
|
22
|
A multidimensional ODE-based model of Alzheimer's disease progression. Sci Rep 2023; 13:3162. [PMID: 36823416 PMCID: PMC9950424 DOI: 10.1038/s41598-023-29383-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 02/03/2023] [Indexed: 02/25/2023] Open
Abstract
Data-driven Alzheimer's disease (AD) progression models are useful for clinical prediction, disease mechanism understanding, and clinical trial design. Most dynamic models were inspired by the amyloid cascade hypothesis and described AD progression as a linear chain of pathological events. However, the heterogeneity observed in healthy and sporadic AD populations challenged the amyloid hypothesis, and there is a need for more flexible dynamical models that accompany this conceptual shift. We present a statistical model of the temporal evolution of biomarkers and cognitive tests that allows diverse biomarker paths throughout the disease. The model consists of two elements: a multivariate dynamic model of the joint evolution of biomarkers and cognitive tests; and a clinical prediction model. The dynamic model uses a system of ordinary differential equations to jointly model the rate of change of an individual's biomarkers and cognitive tests. The clinical prediction model is an ordinal logistic model of the diagnostic label. Prognosis and time-to-onset predictions are obtained by computing the clinical label probabilities throughout the forecasted biomarker trajectories. The proposed dynamical model is interpretable, free of one-dimensional progression hypotheses or disease staging paradigms, and can account for the heterogeneous dynamics observed in sporadic AD. We developed the model using longitudinal data from the Alzheimer's Disease Neuroimaging Initiative. We illustrate the patterns of biomarker rates of change and the model performance to predict the time to conversion from MCI to dementia.
Collapse
|
23
|
Maheux E, Koval I, Ortholand J, Birkenbihl C, Archetti D, Bouteloup V, Epelbaum S, Dufouil C, Hofmann-Apitius M, Durrleman S. Forecasting individual progression trajectories in Alzheimer's disease. Nat Commun 2023; 14:761. [PMID: 36765056 PMCID: PMC9918533 DOI: 10.1038/s41467-022-35712-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 12/19/2022] [Indexed: 02/12/2023] Open
Abstract
The anticipation of progression of Alzheimer's disease (AD) is crucial for evaluations of secondary prevention measures thought to modify the disease trajectory. However, it is difficult to forecast the natural progression of AD, notably because several functions decline at different ages and different rates in different patients. We evaluate here AD Course Map, a statistical model predicting the progression of neuropsychological assessments and imaging biomarkers for a patient from current medical and radiological data at early disease stages. We tested the method on more than 96,000 cases, with a pool of more than 4,600 patients from four continents. We measured the accuracy of the method for selecting participants displaying a progression of clinical endpoints during a hypothetical trial. We show that enriching the population with the predicted progressors decreases the required sample size by 38% to 50%, depending on trial duration, outcome, and targeted disease stage, from asymptomatic individuals at risk of AD to subjects with early and mild AD. We show that the method introduces no biases regarding sex or geographic locations and is robust to missing data. It performs best at the earliest stages of disease and is therefore highly suitable for use in prevention trials.
Collapse
Affiliation(s)
- Etienne Maheux
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Hôpital Pitié-Salpêtrière, Paris, France
| | - Igor Koval
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Hôpital Pitié-Salpêtrière, Paris, France
| | - Juliette Ortholand
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Hôpital Pitié-Salpêtrière, Paris, France
| | - Colin Birkenbihl
- Department of bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, 53115, Germany
| | - Damiano Archetti
- IRCCS Instituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Vincent Bouteloup
- Université de Bordeaux, CNRS UMR 5293, Institut des Maladies Neurodégénératives, Bordeaux, France
- Centre Hospitalier Universitaire (CHU) de Bordeaux, pôle de neurosciences cliniques, centre mémoire de ressources et de recherche, Bordeaux, France
| | - Stéphane Epelbaum
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Hôpital Pitié-Salpêtrière, Institut de la mémoire et de la maladie d'Alzheimer (IM2A), center of excellence of neurodegenerative diseases (CoEN), department of Neurology, DMU Neurosciences, Paris, France
| | - Carole Dufouil
- Université de Bordeaux, CNRS UMR 5293, Institut des Maladies Neurodégénératives, Bordeaux, France
- Centre Hospitalier Universitaire (CHU) de Bordeaux, pôle de neurosciences cliniques, centre mémoire de ressources et de recherche, Bordeaux, France
| | - Martin Hofmann-Apitius
- Department of bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, 53115, Germany
| | - Stanley Durrleman
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Hôpital Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
24
|
Davenport F, Gallacher J, Kourtzi Z, Koychev I, Matthews PM, Oxtoby NP, Parkes LM, Priesemann V, Rowe JB, Smye SW, Zetterberg H. Neurodegenerative disease of the brain: a survey of interdisciplinary approaches. J R Soc Interface 2023; 20:20220406. [PMID: 36651180 PMCID: PMC9846433 DOI: 10.1098/rsif.2022.0406] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 12/16/2022] [Indexed: 01/19/2023] Open
Abstract
Neurodegenerative diseases of the brain pose a major and increasing global health challenge, with only limited progress made in developing effective therapies over the last decade. Interdisciplinary research is improving understanding of these diseases and this article reviews such approaches, with particular emphasis on tools and techniques drawn from physics, chemistry, artificial intelligence and psychology.
Collapse
Affiliation(s)
| | - John Gallacher
- Director of Dementias Platform, Department of Psychiatry, University of Oxford, Oxford, UK
| | - Zoe Kourtzi
- Professor of Cognitive Computational Neuroscience, Department of Psychology, University of Cambridge, UK
| | - Ivan Koychev
- Senior Clinical Researcher, Department of Psychiatry, University of Oxford, Oxford, UK
- Consultant Neuropsychiatrist, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Paul M. Matthews
- Department of Brain Sciences and UK Dementia Research Institute Centre, Imperial College London, Oxford, UK
| | - Neil P. Oxtoby
- UCL Centre for Medical Image Computing and Department of Computer Science, University College London, Gower Street, London, UK
| | - Laura M. Parkes
- School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, UK
| | - Viola Priesemann
- Max Planck Group Leader and Fellow of the Schiemann Kolleg, Max Planck Institute for Dynamics and Self-Organization and Bernstein Center for Computational Neuroscience, Göttingen, Germany
| | - James B. Rowe
- Department of Clinical Neurosciences, MRC Cognition and Brain Sciences Unit and Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
| | | | - Henrik Zetterberg
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, People's Republic of China
| |
Collapse
|
25
|
Koval I, Dighiero-Brecht T, Tobin AJ, Tabrizi SJ, Scahill RI, Tezenas du Montcel S, Durrleman S, Durr A. Forecasting individual progression trajectories in Huntington disease enables more powered clinical trials. Sci Rep 2022; 12:18928. [PMID: 36344508 PMCID: PMC9640581 DOI: 10.1038/s41598-022-18848-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 08/22/2022] [Indexed: 11/09/2022] Open
Abstract
Variability in neurodegenerative disease progression poses great challenges for the evaluation of potential treatments. Identifying the persons who will experience significant progression in the short term is key for the implementation of trials with smaller sample sizes. We apply here disease course mapping to forecast biomarker progression for individual carriers of the pathological CAG repeat expansions responsible for Huntington disease. We used data from two longitudinal studies (TRACK-HD and TRACK-ON) to synchronize temporal progression of 15 clinical and imaging biomarkers from 290 participants with Huntington disease. We used then the resulting HD COURSE MAP to forecast clinical endpoints from the baseline data of 11,510 participants from ENROLL-HD, an external validation cohort. We used such forecasts to select participants at risk for progression and compute the power of trials for such an enriched population. HD COURSE MAP forecasts biomarkers 5 years after the baseline measures with a maximum mean absolute error of 10 points for the total motor score and 2.15 for the total functional capacity. This allowed reducing sample sizes in trial up to 50% including participants with a higher risk for progression ensuring a more homogeneous group of participants.
Collapse
Affiliation(s)
- Igor Koval
- Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, Sorbonne Université, 75013, Paris, France
| | - Thomas Dighiero-Brecht
- Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, Sorbonne Université, 75013, Paris, France
| | - Allan J Tobin
- Biological Adaptation and Ageing, Sorbonne Université, Paris, France
- Brain Research Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Sarah J Tabrizi
- UCL Queen Square Institute of Neurology, University College London, Queen Square, London, UK
| | - Rachael I Scahill
- UCL Queen Square Institute of Neurology, University College London, Queen Square, London, UK
| | - Sophie Tezenas du Montcel
- Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, Sorbonne Université, 75013, Paris, France
| | - Stanley Durrleman
- Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, Sorbonne Université, 75013, Paris, France.
| | - Alexandra Durr
- Department of Neurology, DMU Neurosciences, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, 75013, Paris, France.
| |
Collapse
|
26
|
Redolfi A, Archetti D, De Francesco S, Crema C, Tagliavini F, Lodi R, Ghidoni R, Gandini Wheeler-Kingshott CAM, Alexander DC, D'Angelo E. Italian, European, and international neuroinformatics efforts: An overview. Eur J Neurosci 2022. [PMID: 36310103 DOI: 10.1111/ejn.15854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/18/2022] [Accepted: 10/18/2022] [Indexed: 12/15/2022]
Abstract
Neuroinformatics is a research field that focusses on software tools capable of identifying, analysing, modelling, organising and sharing multiscale neuroscience data. Neuroinformatics has exploded in the last two decades with the emergence of the Big Data phenomenon, characterised by the so-called 3Vs (volume, velocity and variety), which provided neuroscientists with an improved ability to acquire and process data faster and more cheaply thanks to technical improvements in clinical, genomic and radiological technologies. This situation has led to a 'data deluge', as neuroscientists can routinely collect more study data in a few days than they could in a year just a decade ago. To address this phenomenon, several neuroimaging-focussed neuroinformatics platforms have emerged, funded by national or transnational agencies, with the following goals: (i) development of tools for archiving and organising analytical data (XNAT, REDCap and LabKey); (ii) development of data-driven models evolving from reductionist approaches to multidimensional models (RIN, IVN, HBD, EuroPOND, E-DADS and GAAIN BRAIN); and (iii) development of e-infrastructures to provide sufficient computational power and storage resources (neuGRID, HBP-EBRAINS, LONI and CONP). Although the scenario is still fragmented, there are technological and economical attempts at both national and international levels to introduce high standards for open and Findable, Accessible, Interoperable and Reusable (FAIR) neuroscience worldwide.
Collapse
Affiliation(s)
- Alberto Redolfi
- Laboratory of Neuroinformatics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Damiano Archetti
- Laboratory of Neuroinformatics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Silvia De Francesco
- Laboratory of Neuroinformatics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Claudio Crema
- Laboratory of Neuroinformatics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Fabrizio Tagliavini
- Scientific Directorate, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Raffaele Lodi
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Claudia A M Gandini Wheeler-Kingshott
- NMR Research Unit, Queen Square MS Center, Department of Neuroinflammation, UCL Institute of Neurology, London, UK.,Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,Brain Connectivity Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Daniel C Alexander
- Centre for Medical Image Computing, University College London, London, UK.,Department of Computer Science, University College London, London, UK
| | - Egidio D'Angelo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,Brain Connectivity Center, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
27
|
Salimi Y, Domingo-Fernández D, Bobis-Álvarez C, Hofmann-Apitius M, Birkenbihl C. ADataViewer: exploring semantically harmonized Alzheimer's disease cohort datasets. Alzheimers Res Ther 2022; 14:69. [PMID: 35598021 PMCID: PMC9123725 DOI: 10.1186/s13195-022-01009-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 04/27/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Currently, Alzheimer's disease (AD) cohort datasets are difficult to find and lack across-cohort interoperability, and the actual content of publicly available datasets often only becomes clear to third-party researchers once data access has been granted. These aspects severely hinder the advancement of AD research through emerging data-driven approaches such as machine learning and artificial intelligence and bias current data-driven findings towards the few commonly used, well-explored AD cohorts. To achieve robust and generalizable results, validation across multiple datasets is crucial. METHODS We accessed and systematically investigated the content of 20 major AD cohort datasets at the data level. Both, a medical professional and a data specialist, manually curated and semantically harmonized the acquired datasets. Finally, we developed a platform that displays vital information about the available datasets. RESULTS Here, we present ADataViewer, an interactive platform that facilitates the exploration of 20 cohort datasets with respect to longitudinal follow-up, demographics, ethnoracial diversity, measured modalities, and statistical properties of individual variables. It allows researchers to quickly identify AD cohorts that meet user-specified requirements for discovery and validation studies regarding available variables, sample sizes, and longitudinal follow-up. Additionally, we publish the underlying variable mapping catalog that harmonizes 1196 unique variables across the 20 cohorts and paves the way for interoperable AD datasets. CONCLUSIONS In conclusion, ADataViewer facilitates fast, robust data-driven research by transparently displaying cohort dataset content and supporting researchers in selecting datasets that are suited for their envisioned study. The platform is available at https://adata.scai.fraunhofer.de/ .
Collapse
Affiliation(s)
- Yasamin Salimi
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), 53754, Sankt Augustin, Germany.
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115, Bonn, Germany.
| | - Daniel Domingo-Fernández
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), 53754, Sankt Augustin, Germany
| | - Carlos Bobis-Álvarez
- University Hospital Ntra. Sra. de Candelaria, Santa Cruz de Tenerife, 38010, Spain
| | - Martin Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), 53754, Sankt Augustin, Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115, Bonn, Germany
| | - Colin Birkenbihl
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), 53754, Sankt Augustin, Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115, Bonn, Germany
| |
Collapse
|
28
|
Shang Q, Zhang Q, Liu X, Zhu L. Prediction of Early Alzheimer Disease by Hippocampal Volume Changes under Machine Learning Algorithm. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:3144035. [PMID: 35572832 PMCID: PMC9106502 DOI: 10.1155/2022/3144035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/06/2022] [Accepted: 04/15/2022] [Indexed: 11/17/2022]
Abstract
This research was aimed at discussing the application value of different machine learning algorithms in the prediction of early Alzheimer's disease (AD), which was based on hippocampal volume changes in magnetic resonance imaging (MRI). In the research, the 84 cases in American Alzheimer's disease neuroimaging initiative (ADNI) database were selected as the research data. Based on the scoring results of cognitive function, all cases were divided into three groups, including cognitive function normal (normal group), early mild cognitive impairment (e-MCI group), and later mild cognitive impairment (l-MCI group) groups. Each group included 28 cases. The features of hippocampal volume changes in MRI images of the patients in different groups were extracted. The samples of training set and test set were established. Besides, the established support vector machine (SVM), decision tree (DT), and random forest (RF) prediction models were used to predict e-MCI. Metalinear regression was utilized to analyze MRI feature data, and the predictive accuracy, sensitivity, and specificity of different models were calculated. The result showed that the volumes of hippocampal left CA1, left CA2-3, left CA4-DG, left presubiculum, left tail, right CA2-3, right CA4-DG, right presubiculum, and right tail in e-MCI group were all smaller than those in normal group (P < 0.01). The corresponding volume of hippocampal subregions in l-MCI group was remarkably reduced compared with that in normal group (P < 0.001). The volumes of regions left CA1, left CA2-3, left CA4-DG, right CA2-3, right CA4-DG, and right presubiculum were all positively correlated with logical memory test-delay recall (LMT-DR) score (R 2 = 0.1702, 0.3779, 0.1607, 0.1620, 0.0426, and 0.1309; P < 0.001). The predictive accuracy of training set sample by DT, SVM, and RF was 86.67%, 93.33%, and 98.33%, respectively. Based on the changes in the volumes of left CA4-DG, right CA2-3, and right CA4-DG, the predictive accuracy of e-MCI and l-MCI by RF model was both higher than those by DT model (P < 0.01). Besides, the predictive accuracy, sensitivity, and specificity of e-MCI by RF model was all notably higher than those by DT model (P < 0.01). The above results demonstrated that the effective early AD prediction models were established by the volume changes in hippocampal subregions, which was based on RF in the research. The establishment of early AD prediction models offered certain reference basis to the diagnosis and treatment of AD patients.
Collapse
Affiliation(s)
- Qun Shang
- Department of Radiology, Zibo Central Hospital, Zibo, 255000 Shandong, China
| | - Qi Zhang
- Department of Radiology, Zibo Central Hospital, Zibo, 255000 Shandong, China
| | - Xiao Liu
- Department of Radiology, Zibo Central Hospital, Zibo, 255000 Shandong, China
| | - Lingchen Zhu
- Department of Radiology, Zibo Central Hospital, Zibo, 255000 Shandong, China
| |
Collapse
|
29
|
Genetically Targeted Clinical Trials in Parkinson's Disease: Learning from the Successes Made in Oncology. Genes (Basel) 2021; 12:genes12101529. [PMID: 34680924 PMCID: PMC8535305 DOI: 10.3390/genes12101529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/17/2021] [Accepted: 09/24/2021] [Indexed: 12/25/2022] Open
Abstract
Clinical trials in neurodegenerative disorders have been associated with high rate of failures, while in oncology, the implementation of precision medicine and focus on genetically defined subtypes of disease and targets for drug development have seen an unprecedented success. With more than 20 genes associated with Parkinson’s disease (PD), most of which are highly penetrant and often cause early onset or atypical signs and symptoms, and an increasing understanding of the associated pathophysiology culminating in dopaminergic neurodegeneration, applying the technologies and designs into the field of neurodegeneration seems a logical step. This review describes some of the methods used in oncology clinical trials and some attempts in Parkinson’s disease and the potential of further implementing genetics, biomarkers and smart clinical trial designs in this disease area.
Collapse
|
30
|
Ansart M, Epelbaum S, Bassignana G, Bône A, Bottani S, Cattai T, Couronné R, Faouzi J, Koval I, Louis M, Thibeau-Sutre E, Wen J, Wild A, Burgos N, Dormont D, Colliot O, Durrleman S. Predicting the progression of mild cognitive impairment using machine learning: A systematic, quantitative and critical review. Med Image Anal 2020; 67:101848. [PMID: 33091740 DOI: 10.1016/j.media.2020.101848] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 08/17/2020] [Accepted: 08/31/2020] [Indexed: 11/25/2022]
Abstract
We performed a systematic review of studies focusing on the automatic prediction of the progression of mild cognitive impairment to Alzheimer's disease (AD) dementia, and a quantitative analysis of the methodological choices impacting performance. This review included 172 articles, from which 234 experiments were extracted. For each of them, we reported the used data set, the feature types, the algorithm type, performance and potential methodological issues. The impact of these characteristics on the performance was evaluated using a multivariate mixed effect linear regressions. We found that using cognitive, fluorodeoxyglucose-positron emission tomography or potentially electroencephalography and magnetoencephalography variables significantly improved predictive performance compared to not including them, whereas including other modalities, in particular T1 magnetic resonance imaging, did not show a significant effect. The good performance of cognitive assessments questions the wide use of imaging for predicting the progression to AD and advocates for exploring further fine domain-specific cognitive assessments. We also identified several methodological issues, including the absence of a test set, or its use for feature selection or parameter tuning in nearly a fourth of the papers. Other issues, found in 15% of the studies, cast doubts on the relevance of the method to clinical practice. We also highlight that short-term predictions are likely not to be better than predicting that subjects stay stable over time. These issues highlight the importance of adhering to good practices for the use of machine learning as a decision support system for the clinical practice.
Collapse
Affiliation(s)
- Manon Ansart
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France; Inserm, U 1127, Paris, F-75013, France; CNRS, UMR 7225, Paris, F-75013, France; Sorbonne Université, Paris, F-75013, France; Inria, Aramis project-team, Paris, F-75013, France.
| | - Stéphane Epelbaum
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France; Inserm, U 1127, Paris, F-75013, France; CNRS, UMR 7225, Paris, F-75013, France; Sorbonne Université, Paris, F-75013, France; Inria, Aramis project-team, Paris, F-75013, France; Institute of Memory and Alzheimer's Disease (IM2A), Centre of excellence of neurodegenerative disease (CoEN), National Reference Center for Rare or Early Dementias, Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'hôpital, Paris, F-75013, France
| | - Giulia Bassignana
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France; Inserm, U 1127, Paris, F-75013, France; CNRS, UMR 7225, Paris, F-75013, France; Sorbonne Université, Paris, F-75013, France; Inria, Aramis project-team, Paris, F-75013, France
| | - Alexandre Bône
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France; Inserm, U 1127, Paris, F-75013, France; CNRS, UMR 7225, Paris, F-75013, France; Sorbonne Université, Paris, F-75013, France; Inria, Aramis project-team, Paris, F-75013, France
| | - Simona Bottani
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France; Inserm, U 1127, Paris, F-75013, France; CNRS, UMR 7225, Paris, F-75013, France; Sorbonne Université, Paris, F-75013, France; Inria, Aramis project-team, Paris, F-75013, France
| | - Tiziana Cattai
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France; Inserm, U 1127, Paris, F-75013, France; CNRS, UMR 7225, Paris, F-75013, France; Sorbonne Université, Paris, F-75013, France; Inria, Aramis project-team, Paris, F-75013, France; Dept. of Information Engineering, Electronics and Telecommunication, Sapienza University of Rome, Italy
| | - Raphaël Couronné
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France; Inserm, U 1127, Paris, F-75013, France; CNRS, UMR 7225, Paris, F-75013, France; Sorbonne Université, Paris, F-75013, France; Inria, Aramis project-team, Paris, F-75013, France
| | - Johann Faouzi
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France; Inserm, U 1127, Paris, F-75013, France; CNRS, UMR 7225, Paris, F-75013, France; Sorbonne Université, Paris, F-75013, France; Inria, Aramis project-team, Paris, F-75013, France
| | - Igor Koval
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France; Inserm, U 1127, Paris, F-75013, France; CNRS, UMR 7225, Paris, F-75013, France; Sorbonne Université, Paris, F-75013, France; Inria, Aramis project-team, Paris, F-75013, France
| | - Maxime Louis
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France; Inserm, U 1127, Paris, F-75013, France; CNRS, UMR 7225, Paris, F-75013, France; Sorbonne Université, Paris, F-75013, France; Inria, Aramis project-team, Paris, F-75013, France
| | - Elina Thibeau-Sutre
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France; Inserm, U 1127, Paris, F-75013, France; CNRS, UMR 7225, Paris, F-75013, France; Sorbonne Université, Paris, F-75013, France; Inria, Aramis project-team, Paris, F-75013, France
| | - Junhao Wen
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France; Inserm, U 1127, Paris, F-75013, France; CNRS, UMR 7225, Paris, F-75013, France; Sorbonne Université, Paris, F-75013, France; Inria, Aramis project-team, Paris, F-75013, France
| | - Adam Wild
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France; Inserm, U 1127, Paris, F-75013, France; CNRS, UMR 7225, Paris, F-75013, France; Sorbonne Université, Paris, F-75013, France; Inria, Aramis project-team, Paris, F-75013, France
| | - Ninon Burgos
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France; Inserm, U 1127, Paris, F-75013, France; CNRS, UMR 7225, Paris, F-75013, France; Sorbonne Université, Paris, F-75013, France; Inria, Aramis project-team, Paris, F-75013, France
| | - Didier Dormont
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France; Inserm, U 1127, Paris, F-75013, France; CNRS, UMR 7225, Paris, F-75013, France; Sorbonne Université, Paris, F-75013, France; Inria, Aramis project-team, Paris, F-75013, France; AP-HP, Pitié-Salpêtrière hospital, Department of Neuroradiology, Paris, France
| | - Olivier Colliot
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France; Inserm, U 1127, Paris, F-75013, France; CNRS, UMR 7225, Paris, F-75013, France; Sorbonne Université, Paris, F-75013, France; Inria, Aramis project-team, Paris, F-75013, France; Institute of Memory and Alzheimer's Disease (IM2A), Centre of excellence of neurodegenerative disease (CoEN), National Reference Center for Rare or Early Dementias, Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'hôpital, Paris, F-75013, France; AP-HP, Pitié-Salpêtrière hospital, Department of Neuroradiology, Paris, France
| | - Stanley Durrleman
- Inria, Aramis project-team, Paris, F-75013, France; Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France; Inserm, U 1127, Paris, F-75013, France; CNRS, UMR 7225, Paris, F-75013, France; Sorbonne Université, Paris, F-75013, France
| |
Collapse
|