Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Br J Radiol ; 97(1155): 483-491, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38366148

ABSTRACT

Artificial intelligence (AI) methods have been applied to medical imaging for several decades, but in the last few years, the number of publications and the number of AI-enabled medical devices coming on the market have significantly increased. While some AI-enabled approaches are proving very valuable, systematic reviews of the AI imaging field identify significant weaknesses in a significant proportion of the literature. Medical device regulators have recently become more proactive in publishing guidance documents and recognizing standards that will require that the development and validation of AI-enabled medical devices need to be more rigorous than required for tradition "rule-based" software. In particular, developers are required to better identify and mitigate risks (such as bias) that arise in AI-enabled devices, and to ensure that the devices are validated in a realistic clinical setting to ensure their output is clinically meaningful. While this evolving regulatory landscape will mean that device developers will take longer to bring novel AI-based medical imaging devices to market, such additional rigour is necessary to address existing weaknesses in the field and ensure that patients and healthcare professionals can trust AI-enabled devices. There would also be benefits in the academic community taking into account this regulatory framework, to improve the quality of the literature and make it easier for academically developed AI tools to make the transition to medical devices that impact healthcare.


Subject(s)
Artificial Intelligence , Software , Humans , Health Personnel , Publishing
2.
Magn Reson Med ; 86(3): 1194-1211, 2021 09.
Article in English | MEDLINE | ID: mdl-33847012

ABSTRACT

PURPOSE: A standard MRI system phantom has been designed and fabricated to assess scanner performance, stability, comparability and assess the accuracy of quantitative relaxation time imaging. The phantom is unique in having traceability to the International System of Units, a high level of precision, and monitoring by a national metrology institute. Here, we describe the phantom design, construction, imaging protocols, and measurement of geometric distortion, resolution, slice profile, signal-to-noise ratio (SNR), proton-spin relaxation times, image uniformity and proton density. METHODS: The system phantom, designed by the International Society of Magnetic Resonance in Medicine ad hoc committee on Standards for Quantitative MR, is a 200 mm spherical structure that contains a 57-element fiducial array; two relaxation time arrays; a proton density/SNR array; resolution and slice-profile insets. Standard imaging protocols are presented, which provide rapid assessment of geometric distortion, image uniformity, T1 and T2 mapping, image resolution, slice profile, and SNR. RESULTS: Fiducial array analysis gives assessment of intrinsic geometric distortions, which can vary considerably between scanners and correction techniques. This analysis also measures scanner/coil image uniformity, spatial calibration accuracy, and local volume distortion. An advanced resolution analysis gives both scanner and protocol contributions. SNR analysis gives both temporal and spatial contributions. CONCLUSIONS: A standard system phantom is useful for characterization of scanner performance, monitoring a scanner over time, and to compare different scanners. This type of calibration structure is useful for quality assurance, benchmarking quantitative MRI protocols, and to transition MRI from a qualitative imaging technique to a precise metrology with documented accuracy and uncertainty.


Subject(s)
Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Phantoms, Imaging , Signal-To-Noise Ratio
3.
Magn Reson Med ; 79(1): 48-61, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29083101

ABSTRACT

The MRI community is using quantitative mapping techniques to complement qualitative imaging. For quantitative imaging to reach its full potential, it is necessary to analyze measurements across systems and longitudinally. Clinical use of quantitative imaging can be facilitated through adoption and use of a standard system phantom, a calibration/standard reference object, to assess the performance of an MRI machine. The International Society of Magnetic Resonance in Medicine AdHoc Committee on Standards for Quantitative Magnetic Resonance was established in February 2007 to facilitate the expansion of MRI as a mainstream modality for multi-institutional measurements, including, among other things, multicenter trials. The goal of the Standards for Quantitative Magnetic Resonance committee was to provide a framework to ensure that quantitative measures derived from MR data are comparable over time, between subjects, between sites, and between vendors. This paper, written by members of the Standards for Quantitative Magnetic Resonance committee, reviews standardization attempts and then details the need, requirements, and implementation plan for a standard system phantom for quantitative MRI. In addition, application-specific phantoms and implementation of quantitative MRI are reviewed. Magn Reson Med 79:48-61, 2018. © 2017 International Society for Magnetic Resonance in Medicine.


Subject(s)
Brain Mapping/methods , Magnetic Resonance Imaging/methods , Phantoms, Imaging , Algorithms , Biomarkers/metabolism , Calibration , Contrast Media/chemistry , Elasticity , Humans , Image Processing, Computer-Assisted , Linear Models , Models, Theoretical , Perfusion , Reference Values , Reproducibility of Results , Signal-To-Noise Ratio
4.
Neurology ; 87(12): 1235-41, 2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27558378

ABSTRACT

OBJECTIVE: To investigate the effect of enriching mild cognitive impairment (MCI) clinical trials using combined markers of amyloid pathology and neurodegeneration. METHODS: We evaluate an implementation of the recent National Institute for Aging-Alzheimer's Association (NIA-AA) diagnostic criteria for MCI due to Alzheimer disease (AD) as inclusion criteria in clinical trials and assess the effect of enrichment with amyloid (A+), neurodegeneration (N+), and their combination (A+N+) on the rate of clinical progression, required sample sizes, and estimates of trial time and cost. RESULTS: Enrichment based on an individual marker (A+ or N+) substantially improves all assessed trial characteristics. Combined enrichment (A+N+) further improves these results with a reduction in required sample sizes by 45% to 60%, depending on the endpoint. CONCLUSIONS: Operationalizing the NIA-AA diagnostic criteria for clinical trial screening has the potential to substantially improve the statistical power of trials in MCI due to AD by identifying a more rapidly progressing patient population.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/diagnostic imaging , Clinical Trials as Topic , Cognitive Dysfunction/metabolism , Aged , Alzheimer Disease/diagnostic imaging , Biomarkers/cerebrospinal fluid , Brain/metabolism , Clinical Trials as Topic/economics , Clinical Trials as Topic/methods , Cognitive Dysfunction/diagnostic imaging , Cohort Studies , Female , Humans , Male , Mental Status Schedule , Nerve Degeneration/diagnostic imaging , Nerve Degeneration/metabolism , Positron-Emission Tomography
5.
J Magn Reson Imaging ; 41(1): 34-43, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24497105

ABSTRACT

The design and operation of a facility in which a magnetic resonance imaging (MRI) scanner is incorporated into a room used for surgical or endovascular cardiac interventions presents several challenges. MR safety must be maintained in the presence of a much wider variety of equipment than is found in a diagnostic unit, and of staff unfamiliar with the MRI environment, without compromising the safety and practicality of the interventional procedure. Both the MR-guided cardiac interventional unit at Kings College London and the intraoperative imaging suite at the National Hospital for Neurology and Neurosurgery are single-room interventional facilities incorporating 1.5 T cylindrical-bore MRI scanners. The two units employ similar strategies to maintain MR safety, both in original design and day-to-day operational workflows, and between them over a decade of incident-free practice has been accumulated. This article outlines these strategies, highlighting both similarities and differences between the units, as well as some lessons learned and resulting procedural changes made in both units since installation.


Subject(s)
Hospital Departments/organization & administration , Hospital Design and Construction/methods , Magnetic Resonance Imaging, Interventional/methods , Patient Safety , Cardiac Surgical Procedures , Endovascular Procedures , Humans , London , Neurosurgery , United Kingdom
6.
Alzheimers Dement ; 10(4): 421-429.e3, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24985687

ABSTRACT

BACKGROUND: Regulatory qualification of a biomarker for a defined context of use provides scientifically robust assurances to sponsors and regulators that accelerate appropriate adoption of biomarkers into drug development. METHODS: The Coalition Against Major Diseases submitted a dossier to the Scientific Advice Working Party of the European Medicines Agency requesting a qualification opinion on the use of hippocampal volume as a biomarker for enriching clinical trials in subjects with mild cognitive impairment, incorporating a scientific rationale, a literature review and a de novo analysis of Alzheimer's Disease Neuroimaging Initiative data. RESULTS: The literature review and de novo analysis were consistent with the proposed context of use, and the Committee for Medicinal Products for Human Use released an opinion in November 2011. CONCLUSIONS: We summarize the scientific rationale and the data that supported the first qualification of an imaging biomarker by the European Medicines Agency.


Subject(s)
Alzheimer Disease/diagnosis , Alzheimer Disease/therapy , Clinical Trials as Topic , Hippocampus/pathology , Cognitive Dysfunction , Databases, Factual/statistics & numerical data , Disease Progression , Europe , Humans , Neuroimaging , Proportional Hazards Models , ROC Curve
7.
Neurobiol Aging ; 35(4): 808-18, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24211008

ABSTRACT

The objective of this study was to evaluate the effect of computational algorithm, measurement variability, and cut point on hippocampal volume (HCV)-based patient selection for clinical trials in mild cognitive impairment (MCI). We used normal control and amnestic MCI subjects from the Alzheimer's Disease Neuroimaging Initiative 1 (ADNI-1) as normative reference and screening cohorts. We evaluated the enrichment performance of 4 widely used hippocampal segmentation algorithms (FreeSurfer, Hippocampus Multi-Atlas Propagation and Segmentation (HMAPS), Learning Embeddings Atlas Propagation (LEAP), and NeuroQuant) in terms of 2-year changes in Mini-Mental State Examination (MMSE), Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog), and Clinical Dementia Rating Sum of Boxes (CDR-SB). We modeled the implications for sample size, screen fail rates, and trial cost and duration. HCV based patient selection yielded reduced sample sizes (by ∼40%-60%) and lower trial costs (by ∼30%-40%) across a wide range of cut points. These results provide a guide to the choice of HCV cut point for amnestic MCI clinical trials, allowing an informed tradeoff between statistical and practical considerations.


Subject(s)
Algorithms , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/pathology , Hippocampus/pathology , Aged , Aged, 80 and over , Alzheimer Disease , Biomarkers , Clinical Trials as Topic , Female , Humans , Male , Middle Aged , Neuroimaging , Neuropsychological Tests , Patient Selection , Sample Size
8.
Alzheimers Dement ; 7(4): 474-485.e4, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21784356

ABSTRACT

BACKGROUND: The promise of Alzheimer's disease biomarkers has led to their incorporation in new diagnostic criteria and in therapeutic trials; however, significant barriers exist to widespread use. Chief among these is the lack of internationally accepted standards for quantitative metrics. Hippocampal volumetry is the most widely studied quantitative magnetic resonance imaging measure in Alzheimer's disease and thus represents the most rational target for an initial effort at standardization. METHODS AND RESULTS: The authors of this position paper propose a path toward this goal. The steps include the following: (1) Establish and empower an oversight board to manage and assess the effort, (2) adopt the standardized definition of anatomic hippocampal boundaries on magnetic resonance imaging arising from the European Alzheimer's Disease Centers-Alzheimer's Disease Neuroimaging Initiative hippocampal harmonization effort as a reference standard, (3) establish a scientifically appropriate, publicly available reference standard data set based on manual delineation of the hippocampus in an appropriate sample of subjects (Alzheimer's Disease Neuroimaging Initiative), and (4) define minimum technical and prognostic performance metrics for validation of new measurement techniques using the reference standard data set as a benchmark. CONCLUSIONS: Although manual delineation of the hippocampus is the best available reference standard, practical application of hippocampal volumetry will require automated methods. Our intent was to establish a mechanism for credentialing automated software applications to achieve internationally recognized accuracy and prognostic performance standards that lead to the systematic evaluation and then widespread acceptance and use of hippocampal volumetry. The standardization and assay validation process outlined for hippocampal volumetry was envisioned as a template that could be applied to other imaging biomarkers.


Subject(s)
Alzheimer Disease/diagnosis , Hippocampus/pathology , Magnetic Resonance Imaging/methods , Magnetic Resonance Imaging/standards , Aged , Aged, 80 and over , Biomarkers/analysis , Europe , Female , Humans , Image Processing, Computer-Assisted , Male , ROC Curve , Reference Values , Reproducibility of Results
9.
Neuroimage ; 47(4): 1506-13, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19477282

ABSTRACT

Rates of brain atrophy derived from serial magnetic resonance (MR) studies may be used to assess therapies for Alzheimer's disease (AD). These measures may be confounded by changes in scanner voxel sizes. For this reason, the Alzheimer's Disease Neuroimaging Initiative (ADNI) included the imaging of a geometric phantom with every scan. This study compares voxel scaling correction using a phantom with correction using a 9 degrees of freedom (9DOF) registration algorithm. We took 129 pairs of baseline and 1-year repeat scans, and calculated the volume scaling correction, previously measured using the phantom. We used the registration algorithm to quantify any residual scaling errors, and found the algorithm to be unbiased, with no significant (p=0.97) difference between control (n=79) and AD subjects (n=50), but with a mean (SD) absolute volume change of 0.20 (0.20) % due to linear scalings. 9DOF registration was shown to be comparable to geometric phantom correction in terms of the effect on atrophy measurement and unbiased with respect to disease status. These results suggest that the additional expense and logistic effort of scanning a phantom with every patient scan can be avoided by registration-based scaling correction. Furthermore, based upon the atrophy rates in the AD subjects in this study, sample size requirements would be approximately 10-12% lower with (either) correction for voxel scaling than if no correction was used.


Subject(s)
Alzheimer Disease/diagnosis , Artifacts , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Phantoms, Imaging , Subtraction Technique , Algorithms , Cohort Studies , Humans , Magnetic Resonance Imaging/instrumentation , Reproducibility of Results , Sensitivity and Specificity
10.
Brain Res ; 1270: 19-32, 2009 May 13.
Article in English | MEDLINE | ID: mdl-19272356

ABSTRACT

In humans, mutations of amyloid precursor protein (APP) and presenilins (PS) 1 and 2 are associated with amyloid deposition, brain structural change and cognitive decline, like in Alzheimer's disease (AD). Mice expressing these proteins have illuminated neurodegenerative disease processes but, unlike in humans, quantitative imaging has been little used to systematically determine their effects, or those of normal aging, on brain structure in vivo. Accordingly, we investigated wildtype (WT) and TASTPM mice (expressing human APP(695(K595N, M596L)) x PS1(M146V)) longitudinally using MRI. Automated global and local image registration, allied to a standard digital atlas, provided pairwise segmentation of 13 brain regions. We found the mature mouse brain, unlike in humans, enlarges significantly from 6-14 months old (WT 3.8+/-1.7%, mean+/-SD, P<0.0001). Significant changes were also seen in other WT brain regions, providing an anatomical benchmark for comparing other mouse strains and models of brain disorder. In TASTPM, progressive amyloidosis and astrogliosis, detected immunohistochemically, reflected even larger whole brain changes (5.1+/-1.4%, P<0.0001, transgenexage interaction P=0.0311). Normalising regional volumes to whole brain measurements revealed significant, prolonged, WT-TASTPM volume differences, suggesting transgene effects establish at <6 months old of age in most regions. As in humans, gray matter-rich regions decline with age (e.g. thalamus, cerebral cortex and caudoputamen); ventricles and white matter (corpus callosum, corticospinal tract, fornix system) increase; in TASTPMs such trends often varied significantly from WT (especially hippocampus). The pervasive, age-related structural changes between WT and AD transgenic mice (and mouse and human) suggest subtle but fundamental species differences and AD transgene effects.


Subject(s)
Aging/pathology , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Brain/pathology , Presenilin-1/genetics , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/pathology , Amyloidosis/physiopathology , Animals , Disease Models, Animal , Disease Progression , Gliosis/pathology , Gliosis/physiopathology , Immunohistochemistry , Longitudinal Studies , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1/metabolism , Species Specificity , Transgenes/physiology
11.
Neuroimage ; 44(3): 692-700, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19015039

ABSTRACT

The aim of this paper is to investigate techniques that can identify and quantify cross-sectional differences and longitudinal changes in vivo from magnetic resonance images of murine models of brain disease. Two different approaches have been compared. The first approach is a segmentation-based approach: Each subject at each time point is automatically segmented into a number of anatomical structures using atlas-based segmentation. This allows cross-sectional and longitudinal analyses of group differences on a structure-by-structure basis. The second approach is a deformation-based approach: Longitudinal changes are quantified by the registration of each subject's follow-up images to that subject's baseline image. In addition the baseline images can be registered to an atlas allowing voxel-wise analysis of cross-sectional differences between groups. Both approaches have been tested on two groups of mice: A transgenic model of Alzheimer's disease and a wild-type background strain, using serial imaging performed over the age range from 6-14 months. We show that both approaches are able to identify longitudinal and cross-sectional differences. However, atlas-based segmentation suffers from the inability to detect differences across populations and across time in regions which are much smaller than the anatomical regions. In contrast to this, the deformation-based approach can detect statistically significant differences in highly localized areas.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Pattern Recognition, Automated/methods , Subtraction Technique , Algorithms , Animals , Artificial Intelligence , Humans , Imaging, Three-Dimensional/methods , Mice , Reproducibility of Results , Sensitivity and Specificity
12.
Neuroimage ; 44(1): 99-111, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18848632

ABSTRACT

There is great interest in using automatic computational neuroanatomy tools to study ageing and neurodegenerative disease. Voxel-based morphometry (VBM) is one of the most widely used of such techniques. VBM performs voxel-wise statistical analysis of smoothed spatially normalised segmented Magnetic Resonance Images. There are several reasons why the analysis should include only voxels within a certain mask. We show that one of the most commonly used strategies for defining this mask runs a major risk of excluding from the analysis precisely those voxels where the subjects' brains were most vulnerable to atrophy. We investigate the issues related to mask construction, and recommend the use of alternative strategies which greatly decrease this danger of false negatives.


Subject(s)
Alzheimer Disease/pathology , Brain Mapping/methods , Brain/pathology , Image Interpretation, Computer-Assisted/methods , Aged , Aging/pathology , Alzheimer Disease/diagnostic imaging , Atrophy/diagnostic imaging , Atrophy/pathology , Brain/diagnostic imaging , Dementia/diagnostic imaging , Dementia/pathology , Female , Humans , Magnetic Resonance Imaging , Male , Radionuclide Imaging
13.
Neuroimage ; 42(2): 696-709, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18571436

ABSTRACT

The evaluation of atrophy quantification methods based on magnetic resonance imaging have been usually hindered by the lack of realistic gold standard data against which to judge these methods or to help refine them. Recently [Camara, O., Schweiger, M., Scahill, R., Crum, W., Sneller, B., Schnabel, J., Ridgway, G., Cash, D., Hill, D., Fox, N., 2006. Phenomenological model of diffuse global and regional atrophy using finite-element methods. IEEE Trans. Med.l Imaging 25, 1417-1430], we presented a technique in which atrophy is realistically simulated in different tissue compartments or neuroanatomical structures with a phenomenological model. In this study, we have generated a cohort of realistic simulated Alzheimer's disease (AD) images with known amounts of atrophy, mimicking a set of 19 real controls and 27 probable AD subjects, with an improved version of our atrophy simulation methodology. This database was then used to assess the accuracy of several well-known computational anatomy methods which provide global (BSI and SIENA) or local (Jacobian integration) estimates of longitudinal atrophy in brain structures using MR images. SIENA and BSI results correlated very well with gold standard data (Pearson coefficient of 0.962 and 0.969 respectively), achieving small mean absolute differences with respect to the gold standard (percentage change from baseline volume): BSI of 0.23%+/-0.26%; SIENA of 0.22%+/-0.28%. Jacobian integration was guided by both fluid and FFD-based registration techniques and resulting deformation fields and associated Jacobians were compared, region by region, with gold standard ones. The FFD-based technique outperformed the fluid one in all evaluated structures (mean absolute differences from the gold standard in percentage change from baseline volume): whole brain, FFD=0.31%, fluid=0.58%; lateral ventricles, FFD=0.79%; fluid=1.45%; left hippocampus, FFD=0.82%; fluid=1.42%; right hippocampus, FFD=0.95%; fluid=1.62%. The largest errors for both local techniques occurred in the sulcal CSF (FFD=2.27%; fluid=3.55%) regions. For large structures such as the whole brain, these mean absolute differences, relative to the applied atrophy, represented similar percentages for the BSI, SIENA and FFD techniques (controls/patients): BSI, 51.99%/16.36%; SIENA, 62.34%/21.59%; FFD, 41.02%/24.95%. For small structures such as the hippocampi, these percentages were larger, especially for controls where errors were approximately equal to the small applied changes (controls/patients): FFD, 92.82%/43.61%. However, these apparently large relative errors have not prevented the global or hippocampal measures from finding significant group separation in our study. The evaluation framework presented here will help in quantifying whether the accuracy of future methodological developments is sufficient for analysing change in smaller or less atrophied local brain regions. Results obtained in our experiments with realistic simulated data confirm previously published estimates of accuracy for both evaluated global techniques. Regarding Jacobian Integration methods, the FFD-based one demonstrated promising results and potential for being used in clinical studies alongside (or in place of) the more common global methods. The generated gold standard data has also allowed us to identify some stages and sets of parameters in the evaluated techniques--the brain extraction step in the global techniques and the number of multi-resolution levels and the stopping criteria in the registration-based methods--that are critical for their accuracy.


Subject(s)
Aging/pathology , Alzheimer Disease/diagnosis , Brain/pathology , Image Interpretation, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Models, Neurological , Atrophy/diagnosis , Computer Simulation , Humans , Reproducibility of Results , Sensitivity and Specificity
14.
J Cardiovasc Magn Reson ; 10: 13, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18312619

ABSTRACT

A popular technique to reduce respiratory motion for cardiovascular magnetic resonance is to perform a multi-slice acquisition in which a patient holds their breath multiple times during the scan. The feasibility of rigid slice-to-volume registration to correct for misalignments of slice stacks in such images due to differing breath-hold positions is explored. Experimental results indicate that slice-to-volume registration can compensate for the typical misalignments expected. Correction of slice misalignment results in anatomically more correct images, as well as improved left ventricular volume measurements. The interstudy reproducibility has also been improved reducing the number of samples needed for cardiac MR studies.


Subject(s)
Artifacts , Cardiovascular Diseases/diagnosis , Image Interpretation, Computer-Assisted , Imaging, Three-Dimensional , Magnetic Resonance Imaging , Ventricular Function, Left , Algorithms , Cardiovascular Diseases/physiopathology , Computer Simulation , Feasibility Studies , Female , Humans , Male , Models, Cardiovascular , Observer Variation , Predictive Value of Tests , Reproducibility of Results , Respiration
15.
J Magn Reson Imaging ; 27(4): 685-91, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18302232

ABSTRACT

The Alzheimer's Disease Neuroimaging Initiative (ADNI) is a longitudinal multisite observational study of healthy elders, mild cognitive impairment (MCI), and Alzheimer's disease. Magnetic resonance imaging (MRI), (18F)-fluorodeoxyglucose positron emission tomography (FDG PET), urine serum, and cerebrospinal fluid (CSF) biomarkers, as well as clinical/psychometric assessments are acquired at multiple time points. All data will be cross-linked and made available to the general scientific community. The purpose of this report is to describe the MRI methods employed in ADNI. The ADNI MRI core established specifications that guided protocol development. A major effort was devoted to evaluating 3D T(1)-weighted sequences for morphometric analyses. Several options for this sequence were optimized for the relevant manufacturer platforms and then compared in a reduced-scale clinical trial. The protocol selected for the ADNI study includes: back-to-back 3D magnetization prepared rapid gradient echo (MP-RAGE) scans; B(1)-calibration scans when applicable; and an axial proton density-T(2) dual contrast (i.e., echo) fast spin echo/turbo spin echo (FSE/TSE) for pathology detection. ADNI MRI methods seek to maximize scientific utility while minimizing the burden placed on participants. The approach taken in ADNI to standardization across sites and platforms of the MRI protocol, postacquisition corrections, and phantom-based monitoring of all scanners could be used as a model for other multisite trials.


Subject(s)
Alzheimer Disease/diagnosis , Brain/pathology , Magnetic Resonance Imaging/methods , Aged , Alzheimer Disease/pathology , Humans , Magnetic Resonance Imaging/standards
16.
Neuroimage ; 39(4): 1752-62, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18063391

ABSTRACT

Measures of structural brain change based on longitudinal MR imaging are increasingly important but can be degraded by intensity non-uniformity. This non-uniformity can be more pronounced at higher field strengths, or when using multichannel receiver coils. We assessed the ability of the non-parametric non-uniform intensity normalization (N3) technique to correct non-uniformity in 72 volumetric brain MR scans from the preparatory phase of the Alzheimer's Disease Neuroimaging Initiative (ADNI). Normal elderly subjects (n=18) were scanned on different 3-T scanners with a multichannel phased array receiver coil at baseline, using magnetization prepared rapid gradient echo (MP-RAGE) and spoiled gradient echo (SPGR) pulse sequences, and again 2 weeks later. When applying N3, we used five brain masks of varying accuracy and four spline smoothing distances (d=50, 100, 150 and 200 mm) to ascertain which combination of parameters optimally reduces the non-uniformity. We used the normalized white matter intensity variance (standard deviation/mean) to ascertain quantitatively the correction for a single scan; we used the variance of the normalized difference image to assess quantitatively the consistency of the correction over time from registered scan pairs. Our results showed statistically significant (p<0.01) improvement in uniformity for individual scans and reduction in the normalized difference image variance when using masks that identified distinct brain tissue classes, and when using smaller spline smoothing distances (e.g., 50-100 mm) for both MP-RAGE and SPGR pulse sequences. These optimized settings may assist future large-scale studies where 3-T scanners and phased array receiver coils are used, such as ADNI, so that intensity non-uniformity does not influence the power of MR imaging to detect disease progression and the factors that influence it.


Subject(s)
Alzheimer Disease/pathology , Magnetic Resonance Imaging/instrumentation , Aged , Algorithms , Brain/pathology , Calibration , Cognition Disorders/pathology , Data Interpretation, Statistical , Humans , Image Processing, Computer-Assisted , Reproducibility of Results
17.
Med Image Comput Comput Assist Interv ; 10(Pt 2): 785-92, 2007.
Article in English | MEDLINE | ID: mdl-18044640

ABSTRACT

The main goal of this work was to assess the accuracy of several well-known methods which provide global (BSI and SIENA) or local (Jacobian integration) estimates of longitudinal atrophy in brain structures using Magnetic Resonance images. For that purpose, we have generated realistic simulated images which mimic the patterns of change obtained from a cohort of 19 real controls and 27 probable Alzheimer's disease patients. SIENA and BSI results correlate very well with gold standard data (BSI mean absolute error < 0.29%; SIENA < 0.44%). Jacobian integration was guided by both fluid and FFD-based registration techniques and resulting deformation fields and associated Jacobians were compared, region by region, with gold standard ones. The FFD registration technique provided more satisfactory results than the fluid one. Mean absolute error differences between volume changes given by the FFD-based technique and the gold standard were: sulcal CSF < 2.49%; lateral ventricles < 2.25%; brain < 0.36%; hippocampi < 1.42%.


Subject(s)
Algorithms , Alzheimer Disease/diagnosis , Brain/pathology , Image Interpretation, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Pattern Recognition, Automated/methods , Subtraction Technique , Artificial Intelligence , Atrophy/diagnosis , Computer Simulation , Databases, Factual , Humans , Image Enhancement/methods , Longitudinal Studies , Models, Neurological , Reproducibility of Results , Sensitivity and Specificity
18.
Med Phys ; 34(4): 1221-33, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17500454

ABSTRACT

In this paper, we present an evaluation study of a set of registration strategies for the alignment of sequences of 3D dynamic contrast-enhanced magnetic resonance breast images. The accuracy of the optimal registration strategies was determined on unseen data. The evaluation is based on the simulation of physically plausible breast deformations using finite element methods and on contrast-enhanced image pairs without visually detectable motion artifacts. The configuration of the finite element model was chosen according to its ability to predict in vivo breast deformations for two volunteers. We computed transformations for ten patients with 12 simulated deformations each. These deformations were applied to the postcontrast image to model patient motion occurring between pre- and postcontrast image acquisition. The original precontrast images were registered to the corresponding deformed postcontrast images. The performance of several registration configurations (rigid, affine, B-spline based nonrigid, single-resolution, multi-resolution, and volume-preserving) was optimized for five of the ten patients. The images were most accurately aligned with volume-preserving single-resolution nonrigid registration employing 40 or 20 mm control point spacing. When tested on the remaining five patients the optimal configurations reduced the average mean registration error from 1.40 to 0.45 mm for the whole breast tissue and from 1.20 to 0.32 mm for the enhancing lesion. These results were obtained on average within 26 (81) min for 40 (20) mm control point spacing. The visual appearance of the difference images from 30 patients was significantly improved after 20 mm volume-preserving single-resolution nonrigid registration in comparison to no registration or rigid registration. No substantial volume changes within the region of the enhancing lesions were introduced by this nonrigid registration.


Subject(s)
Breast Neoplasms/diagnosis , Breast/pathology , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Magnetic Resonance Imaging/methods , Subtraction Technique , Algorithms , Contrast Media , Female , Humans , Mammography/methods , Reproducibility of Results , Sensitivity and Specificity , Signal Processing, Computer-Assisted
19.
Magn Reson Med ; 57(3): 606-13, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17326164

ABSTRACT

Two-dimensional (2D) breath-hold cine MRI is used to assess cardiac anatomy and function. However, this technique requires cooperation from the patient, and in some cases the scan planning is complicated. Isotropic nonangulated three-dimensional (3D) cardiac MR can overcome some of these problems because it requires minimal planning and can be reformatted in any plane. However, current methods, even those that use undersampling techniques, involve breath-holding for periods that are too long for many patients. Free-breathing respiratory gating sequences represent a possible solution for realizing 3D cine imaging. A real-time respiratory self-gating technique for whole-heart cine MRI is presented. The technique enables assessment of cardiac anatomy and function with minimum planning or patient cooperation. Nonangulated isotropic 3D data were acquired from five healthy volunteers and then reformatted into 2D clinical views. The respiratory self-gating technique is shown to improve image quality in free-breathing scanning. In addition, ventricular volumetric data obtained using the 3D approach were comparable to those acquired with the conventional multislice 2D approach.


Subject(s)
Heart/anatomy & histology , Magnetic Resonance Imaging, Cine/methods , Adult , Algorithms , Artifacts , Electrocardiography , Feasibility Studies , Female , Humans , Imaging, Three-Dimensional , Male , Respiration
20.
IEEE Trans Med Imaging ; 25(12): 1617-26, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17167996

ABSTRACT

Recent innovations in drug therapies have made it highly desirable to obtain sensitive biomarkers of disease progression that can be used to quantify the performance of candidate disease modifying drugs. In order to measure potential image-based biomarkers of disease progression in an experimental model of rheumatoid arthritis (RA), we present two different methods to automatically quantify changes in a bone in in-vivo serial magnetic resonance (MR) images from the model. Both methods are based on rigid and nonrigid image registration to perform the analysis. The first method uses segmentation propagation to delineate a bone from the serial MR images giving a global measure of temporal changes in bone volume. The second method uses rigid body registration to determine intensity change within a bone, and then maps these into a reference coordinate system using nonrigid registration. This gives a local measure of temporal changes in bone lesion volume. We detected significant temporal changes in local bone lesion volume in five out of eight identified candidate bone lesion regions, and significant difference in local bone lesion volume between male and female subjects in three out of eight candidate bone lesion regions. But the global bone volume was found to be fluctuating over time. Finally, we compare our findings with histology of the subjects and the manual segmentation of bone lesions.


Subject(s)
Ankle Joint/pathology , Arthritis, Rheumatoid/pathology , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Pattern Recognition, Automated/methods , Subtraction Technique , Algorithms , Animals , Artificial Intelligence , Disease Progression , Female , Information Storage and Retrieval/methods , Male , Rats , Rats, Inbred Lew , Reproducibility of Results , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...