1
|
Teichmann LSJ, Khalil AA, Villringer K, Fiebach JB, Huwer S, Gibson E, Galinovic I. Evaluation of Siemens Healthineers' StrokeSegApp for automated diffusion and perfusion lesion segmentation in patients with ischemic stroke. Front Neurol 2025; 16:1518477. [PMID: 39926019 PMCID: PMC11804811 DOI: 10.3389/fneur.2025.1518477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/02/2025] [Indexed: 02/11/2025] Open
Abstract
Purpose This study aimed to evaluate the perfomance of Siemens Healthineers' StrokeSegApp performance in automatically segmenting diffusion and perfusion lesions in patients with acute ischemic stroke and to assess its clinical utility in guiding mechanical thrombectomy decisions. Methods This retrospective study used MRI data of acute ischemic stroke patients from the prospective observational single-center 1000Plus study, acquired between September 2008 and June 2013 (clinicaltrials.org; NCT00715533) and manually segmented by radiologists as the ground truth. The performance of the StrokeSegApp was compared against this ground truth using the dice similarity coefficient (DSC) and Bland-Altman plots. The study also evaluated the application's ability to recommend mechanical thrombectomy based on DEFUSE 2 and 3 trial criteria. Results The StrokeSegApp demonstrated a mean DSC of 0.60 (95% CI: 0.57-0.63; n = 241) for diffusion deficit segmentation and 0.80 (95% CI: 0.76-0.85; n = 56) for perfusion deficit segmentation. The mean volume deviation was 0.49 mL for diffusion lesions and -7.69 mL for perfusion lesions. Out of 56 subjects meeting DEFUSE 2/3 criteria in the cohort, it correctly identified mechanical thrombectomy candidates with a sensitivity of 82.1% (95% CI: 63.1-93.9%) and a specificity of 96.4% (95% CI: 81.7-99.9%). Conclusion The Siemens Healthineers' StrokeSegApp provides accurate automated segmentation of ischemic stroke lesions, comparable to human experts as well as similar commercial software, and shows potential as a reliable tool in clinical decision-making for stroke treatment.
Collapse
Affiliation(s)
- Lynnet-Samuel J. Teichmann
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ahmed A. Khalil
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kersten Villringer
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jochen B. Fiebach
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | | | - Eli Gibson
- Siemens Healthineers AG, Erlangen, Germany
| | - Ivana Galinovic
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
2
|
Bani-Sadr A, Hermier M, de Bourguignon C, Mechtouff L, Eker OF, Cappucci M, Tommasino E, Martin A, Cho TH, Derex L, Nighoghossian N, Berthezene Y. Oxygen Extraction Fraction Mapping on Admission Magnetic Resonance Imaging May Predict Recovery of Hyperacute Ischemic Brain Lesions After Successful Thrombectomy: A Retrospective Observational Study. Stroke 2024; 55:2685-2693. [PMID: 39391984 DOI: 10.1161/strokeaha.124.047311] [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: 03/27/2024] [Revised: 08/21/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND In acute stroke, diffusion-weighted imaging (DWI) is used to assess the ischemic core. Dynamic-susceptibility contrast perfusion magnetic resonance imaging allows an estimation of the oxygen extraction fraction (OEF), but the outcome of DWI lesions with increased OEF postrecanalization is unclear. This study investigated the impact of OEF on the fate of DWI lesions in patients achieving recanalization after thrombectomy. METHODS This was a retrospective analysis of the HIBISCUS-STROKE cohort (Cohort of Patients to Identify Biological and Imaging Markers of Cardiovascular Outcomes in Stroke; NCT: 03149705), a single-center observational study that prospectively enrolled patients who underwent magnetic resonance imaging triage for thrombectomy and a day-6 T2-fluid-attenuated inversion recovery (FLAIR) magnetic resonance imaging. Automated postprocessing of admission dynamic-susceptibility contrast perfusion magnetic resonance imaging generated OEF maps. At visual analysis, the OEF status within DWI lesions was assessed in comparison to the contralateral side and correlated with volume changes (difference of ischemic lesion between admission DWI and registered day-6 T2-FLAIR). At voxel-based analysis, recovered DWI regions (lesions present on the admission DWI but absent on the registered day-6 T2-FLAIR) and nonrecovered regions were segmented to extract semiquantitative OEF values. RESULTS Of the participants enrolled from 2016 to 2022, 134 of 321 (41.7%) were included (median age, 71.0 years; 58.2% male; median baseline National Institutes of Health Scale score, 15.0). At visual analysis, 46 of 134 (34.3%) patients had increased OEF within DWI lesions. These patients were more likely to show a reduction in ischemic lesion volumes compared with those without increased OEF (median change, -4.0 versus 4.8 mL; P<0.0001). Multivariable analysis indicated that increased OEF within DWI lesions was associated with a reduction in ischemic lesion volumes from admission DWI to day-6 T2-FLAIR (odds ratio, 0.68 [95% CI, 0.49-0.87]; P=0.008). At voxel-based analysis, recovered DWI regions had increased OEF, while nonrecovered regions had decreased OEF (median, 126.9% versus -27.0%; P<0.0001). CONCLUSIONS Increased OEF within hyperacute DWI lesions was associated with ischemic lesion recovery between admission DWI and day-6 T2-FLAIR in patients achieving recanalization after thrombectomy. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT03149705.
Collapse
Affiliation(s)
- Alexandre Bani-Sadr
- Department of Neuroradiology (A.B.-S., M.H., O.F.E., M.C., E.T., A.M., Y.B.), East Group Hospital, Hospices Civils de Lyon, Bron, France
- Centre de Recherche et de Traitement de l'Image pour la Santé (CREATIS) Laboratory, Centre National de la Recherche en Santé (CNRS) Unité Mixte de Recherche (UMR) 5220, Institut National de la Santé et de la Recherche Médicale (INSERM) U1294, Claude Bernard Lyon I University, Villeurbanne, France (A.B.-S., O.F.E., E.T., A.M., Y.B.)
| | - Marc Hermier
- Department of Neuroradiology (A.B.-S., M.H., O.F.E., M.C., E.T., A.M., Y.B.), East Group Hospital, Hospices Civils de Lyon, Bron, France
| | | | - Laura Mechtouff
- Stroke Department (L.M., T.-H.C., L.D., N.N.), East Group Hospital, Hospices Civils de Lyon, Bron, France
- CarMeN Laboratory, INSERM U1060/INRA U1397, Claude Bernard Lyon I University, Bron, France (L.M., T.-H.C., N.N.)
| | - Omer F Eker
- Department of Neuroradiology (A.B.-S., M.H., O.F.E., M.C., E.T., A.M., Y.B.), East Group Hospital, Hospices Civils de Lyon, Bron, France
- Centre de Recherche et de Traitement de l'Image pour la Santé (CREATIS) Laboratory, Centre National de la Recherche en Santé (CNRS) Unité Mixte de Recherche (UMR) 5220, Institut National de la Santé et de la Recherche Médicale (INSERM) U1294, Claude Bernard Lyon I University, Villeurbanne, France (A.B.-S., O.F.E., E.T., A.M., Y.B.)
| | - Matteo Cappucci
- Department of Neuroradiology (A.B.-S., M.H., O.F.E., M.C., E.T., A.M., Y.B.), East Group Hospital, Hospices Civils de Lyon, Bron, France
| | - Emanuele Tommasino
- Department of Neuroradiology (A.B.-S., M.H., O.F.E., M.C., E.T., A.M., Y.B.), East Group Hospital, Hospices Civils de Lyon, Bron, France
- Centre de Recherche et de Traitement de l'Image pour la Santé (CREATIS) Laboratory, Centre National de la Recherche en Santé (CNRS) Unité Mixte de Recherche (UMR) 5220, Institut National de la Santé et de la Recherche Médicale (INSERM) U1294, Claude Bernard Lyon I University, Villeurbanne, France (A.B.-S., O.F.E., E.T., A.M., Y.B.)
| | - Anna Martin
- Department of Neuroradiology (A.B.-S., M.H., O.F.E., M.C., E.T., A.M., Y.B.), East Group Hospital, Hospices Civils de Lyon, Bron, France
- Centre de Recherche et de Traitement de l'Image pour la Santé (CREATIS) Laboratory, Centre National de la Recherche en Santé (CNRS) Unité Mixte de Recherche (UMR) 5220, Institut National de la Santé et de la Recherche Médicale (INSERM) U1294, Claude Bernard Lyon I University, Villeurbanne, France (A.B.-S., O.F.E., E.T., A.M., Y.B.)
| | - Tae-Hee Cho
- Stroke Department (L.M., T.-H.C., L.D., N.N.), East Group Hospital, Hospices Civils de Lyon, Bron, France
- CarMeN Laboratory, INSERM U1060/INRA U1397, Claude Bernard Lyon I University, Bron, France (L.M., T.-H.C., N.N.)
| | - Laurent Derex
- Stroke Department (L.M., T.-H.C., L.D., N.N.), East Group Hospital, Hospices Civils de Lyon, Bron, France
| | - Nobert Nighoghossian
- Stroke Department (L.M., T.-H.C., L.D., N.N.), East Group Hospital, Hospices Civils de Lyon, Bron, France
- CarMeN Laboratory, INSERM U1060/INRA U1397, Claude Bernard Lyon I University, Bron, France (L.M., T.-H.C., N.N.)
| | - Yves Berthezene
- Department of Neuroradiology (A.B.-S., M.H., O.F.E., M.C., E.T., A.M., Y.B.), East Group Hospital, Hospices Civils de Lyon, Bron, France
- Centre de Recherche et de Traitement de l'Image pour la Santé (CREATIS) Laboratory, Centre National de la Recherche en Santé (CNRS) Unité Mixte de Recherche (UMR) 5220, Institut National de la Santé et de la Recherche Médicale (INSERM) U1294, Claude Bernard Lyon I University, Villeurbanne, France (A.B.-S., O.F.E., E.T., A.M., Y.B.)
| |
Collapse
|
3
|
Varatharaj A, Jacob C, Darekar A, Yuen B, Cramer S, Larsson H, Galea I. Measurement variability of blood-brain barrier permeability using dynamic contrast-enhanced magnetic resonance imaging. IMAGING NEUROSCIENCE (CAMBRIDGE, MASS.) 2024; 2:1-16. [PMID: 39449749 PMCID: PMC11497077 DOI: 10.1162/imag_a_00324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/15/2024] [Accepted: 09/11/2024] [Indexed: 10/26/2024]
Abstract
Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) is used to quantify the blood-brain barrier (BBB) permeability-surface area product. Serial measurements can indicate changes in BBB health, of interest to the study of normal physiology, neurological disease, and the effect of therapeutics. We performed a scan-rescan study to inform both sample size calculation for future studies and an appropriate reference change value for patient care. The final dataset included 28 healthy individuals (mean age 53.0 years, 82% female) scanned twice with mean interval 9.9 weeks. DCE-MRI was performed at 3T using a 3D gradient echo sequence with whole brain coverage, T1 mapping using variable flip angles, and a 16-min dynamic sequence with a 3.2-s time resolution. Segmentation of white and grey matter (WM/GM) was performed using a 3D magnetization-prepared gradient echo image. The influx constant Ki was calculated using the Patlak method. The primary outcome was the within-subject coefficient of variation (CV) of Ki in both WM and GM. Ki values followed biological expectations in relation to known GM/WM differences in cerebral blood volume (CBV) and consequently vascular surface area. Subject-derived arterial input functions showed marked within-subject variability which were significantly reduced by using a venous input function (CV of area under the curve 46 vs. 12%, p < 0.001). Use of the venous input function significantly improved the CV of Ki in both WM (30 vs. 59%, p < 0.001) and GM (21 vs. 53%, p < 0.001). Further improvement was obtained using motion correction, scaling the venous input function by the artery, and using the median rather than the mean of individual voxel data. The final method gave CV of 27% and 17% in WM and GM, respectively. No further improvement was obtained by replacing the subject-derived input function by one standard population input function. CV of Ki was shown to be highly sensitive to dynamic sequence duration, with shorter measurement periods giving marked deterioration especially in WM. In conclusion, measurement variability of 3D brain DCE-MRI is sensitive to analysis method and a large precision improvement is obtained using a venous input function.
Collapse
Affiliation(s)
- Aravinthan Varatharaj
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Carmen Jacob
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Angela Darekar
- Medical Physics, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Brian Yuen
- Medical Statistics, Primary Care, Population Sciences and Medical Education, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Stig Cramer
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, Glostrup, Denmark
| | - Henrik Larsson
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, Glostrup, Denmark
| | - Ian Galea
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| |
Collapse
|
4
|
Aamand R, Rasmussen PM, Andersen KS, de Paoli S, Weitzberg E, Christiansen M, Lund TE, Østergaard L. Cerebral microvascular changes in healthy carriers of the APOE-ɛ4 Alzheimer's disease risk gene. PNAS NEXUS 2024; 3:pgae369. [PMID: 39253395 PMCID: PMC11382292 DOI: 10.1093/pnasnexus/pgae369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024]
Abstract
APOE-ɛ4 is a genetic risk factor for Alzheimer's disease (AD). AD is associated with reduced cerebral blood flow (CBF) and with microvascular changes that limit the transport of oxygen from blood into brain tissue: reduced microvascular cerebral blood volume and high relative transit time heterogeneity (RTH). Healthy APOE-ɛ4 carriers reveal brain regions with elevated CBF compared with carriers of the common ɛ3 allele. Such asymptomatic hyperemia may reflect microvascular dysfunction: a vascular disease entity characterized by suboptimal tissue oxygen uptake, rather than limited blood flow per se. Here, we used perfusion MRI to show that elevated regional CBF is accompanied by reduced capillary blood volume in healthy APOE-ɛ4 carriers (carriers) aged 30-70 years compared with similarly aged APOE-ɛ3 carriers (noncarriers). Younger carriers have elevated hippocampal RTH and more extreme RTH values throughout both white matter (WM) and cortical gray matter (GM) compared with noncarriers. Older carriers have reduced WM CBF and more extreme GM RTH values than noncarriers. Across all groups, lower WM and hippocampal RTH correlate with higher educational attainment, which is associated with lower AD risk. Three days of dietary nitrate supplementation increased carriers' WM CBF but caused older carriers to score worse on two of six aggregate neuropsychological scores. The intervention improved late recall in younger carriers and in noncarriers. The APOE-ɛ4 gene is associated with microvascular changes that may impair tissue oxygen extraction. We speculate that vascular risk factor control is particularly important for APOE-ɛ4 carriers' healthy aging.
Collapse
Affiliation(s)
- Rasmus Aamand
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Peter M Rasmussen
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Katrine Schilling Andersen
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Stine de Paoli
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
- Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Michael Christiansen
- Department for Congenital Disorders, Statens Serum Institut, 2300 Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Torben E Lund
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Leif Østergaard
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
- Department of Neuroradiology, Aarhus University Hospital, 8200 Aarhus, Denmark
| |
Collapse
|
5
|
Foltyn-Dumitru M, Kessler T, Sahm F, Wick W, Heiland S, Bendszus M, Vollmuth P, Schell M. Cluster-based prognostication in glioblastoma: Unveiling heterogeneity based on diffusion and perfusion similarities. Neuro Oncol 2024; 26:1099-1108. [PMID: 38153923 PMCID: PMC11145444 DOI: 10.1093/neuonc/noad259] [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: 09/10/2023] [Indexed: 12/30/2023] Open
Abstract
BACKGROUND While the association between diffusion and perfusion magnetic resonance imaging (MRI) and survival in glioblastoma is established, prognostic models for patients are lacking. This study employed clustering of functional imaging to identify distinct functional phenotypes in untreated glioblastomas, assessing their prognostic significance for overall survival. METHODS A total of 289 patients with glioblastoma who underwent preoperative multimodal MR imaging were included. Mean values of apparent diffusion coefficient normalized relative cerebral blood volume and relative cerebral blood flow were calculated for different tumor compartments and the entire tumor. Distinct imaging patterns were identified using partition around medoids (PAM) clustering on the training dataset, and their ability to predict overall survival was assessed. Additionally, tree-based machine-learning models were trained to ascertain the significance of features pertaining to cluster membership. RESULTS Using the training dataset (231/289) we identified 2 stable imaging phenotypes through PAM clustering with significantly different overall survival (OS). Validation in an independent test set revealed a high-risk group with a median OS of 10.2 months and a low-risk group with a median OS of 26.6 months (P = 0.012). Patients in the low-risk cluster had high diffusion and low perfusion values throughout, while the high-risk cluster displayed the reverse pattern. Including cluster membership in all multivariate Cox regression analyses improved performance (P ≤ 0.004 each). CONCLUSIONS Our research demonstrates that data-driven clustering can identify clinically relevant, distinct imaging phenotypes, highlighting the potential role of diffusion, and perfusion MRI in predicting survival rates of glioblastoma patients.
Collapse
Affiliation(s)
- Martha Foltyn-Dumitru
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
- Section for Computational Neuroimaging, Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Tobias Kessler
- Department of Neurology and Neurooncology Program, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felix Sahm
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Wolfgang Wick
- Department of Neurology and Neurooncology Program, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sabine Heiland
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Philipp Vollmuth
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
- Section for Computational Neuroimaging, Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marianne Schell
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
- Section for Computational Neuroimaging, Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
6
|
Oh G, Moon Y, Moon WJ, Ye JC. Unpaired deep learning for pharmacokinetic parameter estimation from dynamic contrast-enhanced MRI without AIF measurements. Neuroimage 2024; 291:120571. [PMID: 38518829 DOI: 10.1016/j.neuroimage.2024.120571] [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: 11/23/2023] [Revised: 02/28/2024] [Accepted: 03/08/2024] [Indexed: 03/24/2024] Open
Abstract
DCE-MRI provides information about vascular permeability and tissue perfusion through the acquisition of pharmacokinetic parameters. However, traditional methods for estimating these pharmacokinetic parameters involve fitting tracer kinetic models, which often suffer from computational complexity and low accuracy due to noisy arterial input function (AIF) measurements. Although some deep learning approaches have been proposed to tackle these challenges, most existing methods rely on supervised learning that requires paired input DCE-MRI and labeled pharmacokinetic parameter maps. This dependency on labeled data introduces significant time and resource constraints and potential noise in the labels, making supervised learning methods often impractical. To address these limitations, we present a novel unpaired deep learning method for estimating pharmacokinetic parameters and the AIF using a physics-driven CycleGAN approach. Our proposed CycleGAN framework is designed based on the underlying physics model, resulting in a simpler architecture with a single generator and discriminator pair. Crucially, our experimental results indicate that our method does not necessitate separate AIF measurements and produces more reliable pharmacokinetic parameters than other techniques.
Collapse
Affiliation(s)
- Gyutaek Oh
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291, Daehak-ro, Yuseong-gu, 34141, Daejeon, Republic of Korea
| | - Yeonsil Moon
- Department of Neurology, Konkuk University Medical Center, 120-1, Neungdong-ro, Gwangjin-gu, 05030, Seoul, Republic of Korea
| | - Won-Jin Moon
- Department of Radiology, Konkuk University Medical Center, 120-1, Neungdong-ro, Gwangjin-gu, 05030, Seoul, Republic of Korea.
| | - Jong Chul Ye
- Kim Jaechul Graduate School of AI, Korea Advanced Institute of Science and Technology (KAIST), 291, Daehak-ro, Yuseong-gu, 34141, Daejeon, Republic of Korea.
| |
Collapse
|
7
|
van Houdt PJ, Ragunathan S, Berks M, Ahmed Z, Kershaw LE, Gurney-Champion OJ, Tadimalla S, Arvidsson J, Sun Y, Kallehauge J, Dickie B, Lévy S, Bell L, Sourbron S, Thrippleton MJ. Contrast-agent-based perfusion MRI code repository and testing framework: ISMRM Open Science Initiative for Perfusion Imaging (OSIPI). Magn Reson Med 2024; 91:1774-1786. [PMID: 37667526 DOI: 10.1002/mrm.29826] [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: 03/24/2023] [Revised: 06/30/2023] [Accepted: 07/25/2023] [Indexed: 09/06/2023]
Abstract
PURPOSE Software has a substantial impact on quantitative perfusion MRI values. The lack of generally accepted implementations, code sharing and transparent testing reduces reproducibility, hindering the use of perfusion MRI in clinical trials. To address these issues, the ISMRM Open Science Initiative for Perfusion Imaging (OSIPI) aimed to establish a community-led, centralized repository for sharing open-source code for processing contrast-based perfusion imaging, incorporating an open-source testing framework. METHODS A repository was established on the OSIPI GitHub website. Python was chosen as the target software language. Calls for code contributions were made to OSIPI members, the ISMRM Perfusion Study Group, and publicly via OSIPI websites. An automated unit-testing framework was implemented to evaluate the output of code contributions, including visual representation of the results. RESULTS The repository hosts 86 implementations of perfusion processing steps contributed by 12 individuals or teams. These cover all core aspects of DCE- and DSC-MRI processing, including multiple implementations of the same functionality. Tests were developed for 52 implementations, covering five analysis steps. For T1 mapping, signal-to-concentration conversion and population AIF functions, different implementations resulted in near-identical output values. For the five pharmacokinetic models tested (Tofts, extended Tofts-Kety, Patlak, two-compartment exchange, and two-compartment uptake), differences in output parameters were observed between contributions. CONCLUSIONS The OSIPI DCE-DSC code repository represents a novel community-led model for code sharing and testing. The repository facilitates the re-use of existing code and the benchmarking of new code, promoting enhanced reproducibility in quantitative perfusion imaging.
Collapse
Affiliation(s)
- Petra J van Houdt
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Michael Berks
- Quantitative Biomedical Imaging Laboratory, Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Zaki Ahmed
- Corewell Health William Beaumont University Hospital, Diagnostic Radiology, Royal Oak, USA
| | - Lucy E Kershaw
- Edinburgh Imaging and Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Oliver J Gurney-Champion
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Sirisha Tadimalla
- Institute of Medical Physics, The University of Sydney, Sydney, Australia
| | - Jonathan Arvidsson
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Yu Sun
- Institute of Medical Physics, The University of Sydney, Sydney, Australia
| | - Jesper Kallehauge
- Aarhus University Hospital, Danish Centre for Particle Therapy, Aarhus, Denmark
- Aarhus University, Department of Clinical Medicine, Aarhus, Denmark
| | - Ben Dickie
- Division of Informatics, Imaging, and Data Science, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, The University of Manchester, Manchester, UK
| | - Simon Lévy
- MR Research Collaborations, Siemens Healthcare Pty Ltd, Melbourne, Australia
| | - Laura Bell
- Genentech, Inc, Clinical Imaging Group, South San Francisco, USA
| | - Steven Sourbron
- University of Sheffield, Department of Infection, Immunity and Cardiovascular Disease, Sheffield, UK
| | - Michael J Thrippleton
- University of Edinburgh, Edinburgh Imaging and Centre for Clinical Brain Sciences, Edinburgh, UK
| |
Collapse
|
8
|
Tseng CH, Jaspers J, Romero AM, Wielopolski P, Smits M, van Osch MJP, Vos F. Improved reliability of perfusion estimation in dynamic susceptibility contrast MRI by using the arterial input function from dynamic contrast enhanced MRI. NMR IN BIOMEDICINE 2024; 37:e5038. [PMID: 37712359 DOI: 10.1002/nbm.5038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 08/02/2023] [Accepted: 08/23/2023] [Indexed: 09/16/2023]
Abstract
The arterial input function (AIF) plays a crucial role in estimating quantitative perfusion properties from dynamic susceptibility contrast (DSC) MRI. An important issue, however, is that measuring the AIF in absolute contrast-agent concentrations is challenging, due to uncertainty in relation to the measuredR 2 ∗ -weighted signal, signal depletion at high concentration, and partial-volume effects. A potential solution could be to derive the AIF from separately acquired dynamic contrast enhanced (DCE) MRI data. We aim to compare the AIF determined from DCE MRI with the AIF from DSC MRI, and estimated perfusion coefficients derived from DSC data using a DCE-driven AIF with perfusion coefficients determined using a DSC-based AIF. AIFs were manually selected in branches of the middle cerebral artery (MCA) in both DCE and DSC data in each patient. In addition, a semi-automatic AIF-selection algorithm was applied to the DSC data. The amplitude and full width at half-maximum of the AIFs were compared statistically using the Wilcoxon rank-sum test, applying a 0.05 significance level. Cerebral blood flow (CBF) was derived with different AIF approaches and compared further. The results showed that the AIFs extracted from DSC scans yielded highly variable peaks across arteries within the same patient. The semi-automatic DSC-AIF had significantly narrower width compared with the manual AIFs, and a significantly larger peak than the manual DSC-AIF. Additionally, the DCE-based AIF provided a more stable measurement of relative CBF and absolute CBF values estimated with DCE-AIFs that were compatible with previously reported values. In conclusion, DCE-based AIFs were reproduced significantly better across vessels, showed more realistic profiles, and delivered more stable and reasonable CBF measurements. The DCE-AIF can, therefore, be considered as an alternative AIF source for quantitative perfusion estimations in DSC MRI.
Collapse
Affiliation(s)
- Chih-Hsien Tseng
- Department of Imaging Physics, Delft University of Technology, Delft, the Netherlands
- Medical Delta, Delft, the Netherlands
- Holland Proton Therapy Center Consortium-Erasmus MC, Rotterdam, Holland Proton Therapy Centre, Delft, Leiden University Medical Center, Leiden and Delft University of Technology, Delft, the Netherlands
| | - Jaap Jaspers
- Holland Proton Therapy Center Consortium-Erasmus MC, Rotterdam, Holland Proton Therapy Centre, Delft, Leiden University Medical Center, Leiden and Delft University of Technology, Delft, the Netherlands
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Alejandra Mendez Romero
- Holland Proton Therapy Center Consortium-Erasmus MC, Rotterdam, Holland Proton Therapy Centre, Delft, Leiden University Medical Center, Leiden and Delft University of Technology, Delft, the Netherlands
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Piotr Wielopolski
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marion Smits
- Medical Delta, Delft, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Brain Tumour Center, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Matthias J P van Osch
- Medical Delta, Delft, the Netherlands
- Holland Proton Therapy Center Consortium-Erasmus MC, Rotterdam, Holland Proton Therapy Centre, Delft, Leiden University Medical Center, Leiden and Delft University of Technology, Delft, the Netherlands
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Frans Vos
- Department of Imaging Physics, Delft University of Technology, Delft, the Netherlands
- Medical Delta, Delft, the Netherlands
- Holland Proton Therapy Center Consortium-Erasmus MC, Rotterdam, Holland Proton Therapy Centre, Delft, Leiden University Medical Center, Leiden and Delft University of Technology, Delft, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
9
|
Kurokawa R, Kurokawa M, Baba A, Kim J, Srinivasan A, Moritani T. Dynamic susceptibility contrast perfusion-weighted and diffusion-weighted magnetic resonance imaging findings in pilocytic astrocytoma and H3.3 and H3.1 variant diffuse midline glioma, H3K27-altered. PLoS One 2023; 18:e0288412. [PMID: 37450487 PMCID: PMC10348548 DOI: 10.1371/journal.pone.0288412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 06/26/2023] [Indexed: 07/18/2023] Open
Abstract
OBJECTIVE This study compared the dynamic susceptibility contrast (DSC) magnetic resonance imaging parameters and apparent diffusion coefficient (ADC) between pilocytic astrocytoma (PA) and diffuse midline glioma, H3K27-altered (DMG) variants. METHODS The normalized relative cerebral blood volume (nrCBV), normalized relative flow (nrCBF), percentile signal recovery (PSR), and normalized mean ADC (nADCmean) of 23 patients with midline PAs (median age, 13 years [range, 1-71 years]; 13 female patients) and 40 patients with DMG (8.5 years [1-35 years]; 19 female patients), including 35 patients with H3.3- and five patients with H3.1-mutant tumors, treated between January 2016 and May 2022 were statistically compared. RESULTS DMG had a significantly lower nADCmean (median: 1.48 vs. 1.96; p = 0.00075) and lower PSR (0.97 vs. 1.23, p = 0.13) but higher nrCBV and nrCBF (1.66 vs. 1.17, p = 0.058, respectively, and 1.87 vs. 1.19, p = 0.028, respectively) than PA. The H3.3 variant had a lower nADCmean than the H3.1 variant (1.46 vs. 1.80, p = 0.10). CONCLUSION DMG had lower ADC and PSR and higher rCBV and rCBF than PA. The H3.3 variant had a lower ADC than the H3.1 variant. Recognizing the differences and similarities in the DSC parameters and ADC between these tumors may help presurgical diagnosis.
Collapse
Affiliation(s)
- Ryo Kurokawa
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, MI, United States of America
| | - Mariko Kurokawa
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, MI, United States of America
| | - Akira Baba
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, MI, United States of America
| | - John Kim
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, MI, United States of America
| | - Ashok Srinivasan
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, MI, United States of America
| | - Toshio Moritani
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, MI, United States of America
| |
Collapse
|
10
|
Takamura T, Hara S, Nariai T, Ikenouchi Y, Suzuki M, Taoka T, Ida M, Ishigame K, Hori M, Sato K, Kamagata K, Kumamaru K, Oishi H, Okamoto S, Araki Y, Uda K, Miyajima M, Maehara T, Inaji M, Tanaka Y, Naganawa S, Kawai H, Nakane T, Tsurushima Y, Onodera T, Nojiri S, Aoki S. Effect of Temporal Sampling Rate on Estimates of the Perfusion Parameters for Patients with Moyamoya Disease Assessed with Simultaneous Multislice Dynamic Susceptibility Contrast-enhanced MR Imaging. Magn Reson Med Sci 2023; 22:301-312. [PMID: 35296610 PMCID: PMC10449549 DOI: 10.2463/mrms.mp.2021-0162] [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: 12/14/2021] [Accepted: 02/19/2022] [Indexed: 11/09/2022] Open
Abstract
PURPOSE The effect of temporal sampling rate (TSR) on perfusion parameters has not been fully investigated in Moyamoya disease (MMD); therefore, this study evaluated the influence of different TSRs on perfusion parameters quantitatively and qualitatively by applying simultaneous multi-slice (SMS) dynamic susceptibility contrast-enhanced MR imaging (DSC-MRI). METHODS DSC-MRI datasets were acquired from 28 patients with MMD with a TSR of 0.5 s. Cerebral blood flow (CBF), cerebral blood volume (CBV), mean transit time (MTT), time to peak (TTP), and time to maximum tissue residue function (Tmax) were calculated for eight TSRs ranging from 0.5 to 4.0 s in 0.5-s increments that were subsampled from a TSR of 0.5 s datasets. Perfusion measurements and volume for chronic ischemic (Tmax ≥ 2 s) and non-ischemic (Tmax < 2 s) areas for each TSR were compared to measurements with a TSR of 0.5 s, as was visual perfusion map analysis. RESULTS CBF, CBV, and Tmax values tended to be underestimated, whereas MTT and TTP values were less influenced, with a longer TSR. Although Tmax values were overestimated in the TSR of 1.0 s in non-ischemic areas, differences in perfusion measurements between the TSRs of 0.5 and 1.0 s were generally minimal. The volumes of the chronic ischemic areas with a TSR ≥ 3.0 s were significantly underestimated. In CBF and CBV maps, no significant deterioration was noted in image quality up to 3.0 and 2.5 s, respectively. The image quality of MTT, TTP, and Tmax maps for the TSR of 1.0 s was similar to that for the TSR of 0.5 s but was significantly deteriorated for the TSRs of ≥ 1.5 s. CONCLUSION In the assessment of MMD by SMS DSC-MRI, application of TSRs of ≥ 1.5 s may lead to deterioration of the perfusion measurements; however, that was less influenced in TSRs of ≤ 1.0 s.
Collapse
Affiliation(s)
- Tomohiro Takamura
- Department of Radiology, Shizuoka General Hospital, Shizuoka, Shizuoka, Japan
- Department of Radiology, Juntendo University, Tokyo, Japan
| | - Shoko Hara
- Department of Neurosurgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tadashi Nariai
- Department of Neurosurgery, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | - Toshiaki Taoka
- Department of Radiology, Nagoya University, Nagoya, Aichi, Japan
| | - Masahiro Ida
- Department of Radiology, Mito Medical Center, Higashiibaraki, Ibaraki, Japan
| | - Keiichi Ishigame
- Department of Radiology, Kenshinkai Tokyo Medical Clinic, Tokyo, Japan
| | - Masaaki Hori
- Department of Radiology, Juntendo University, Tokyo, Japan
- Department of Radiology, Toho University Omori Medical Center, Tokyo, Japan
| | - Kanako Sato
- Department of Radiology, Juntendo University, Tokyo, Japan
| | - Koji Kamagata
- Department of Radiology, Juntendo University, Tokyo, Japan
| | | | - Hidenori Oishi
- Department of Neurosurgery, Juntendo University, Tokyo, Japan
| | - Sho Okamoto
- Department of Neurosurgery, Nagoya University, Nagoya, Aichi, Japan
| | - Yoshio Araki
- Department of Neurosurgery, Nagoya University, Nagoya, Aichi, Japan
| | - Kenji Uda
- Department of Neurosurgery, Nagoya University, Nagoya, Aichi, Japan
| | - Masakazu Miyajima
- Department of Neurosurgery, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo, Japan
| | - Taketoshi Maehara
- Department of Neurosurgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Motoki Inaji
- Department of Neurosurgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoji Tanaka
- Department of Neurosurgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinji Naganawa
- Department of Radiology, Nagoya University, Nagoya, Aichi, Japan
| | - Hisashi Kawai
- Department of Radiology, Nagoya University, Nagoya, Aichi, Japan
| | - Toshiki Nakane
- Department of Radiology, Nagoya University, Nagoya, Aichi, Japan
| | | | - Toshiyuki Onodera
- Department of Radiology, Tokyo Metropolitan Cancer Detection Center, Tokyo, Japan
| | - Shuko Nojiri
- Clinical Research and Trial Center, Juntendo Hospital, Tokyo, Japan
| | - Shigeki Aoki
- Department of Radiology, Juntendo University, Tokyo, Japan
| |
Collapse
|
11
|
Ota Y, Liao E, Zhao R, Capizzano AA, Baba A, Lobo R, Shah G, Srinivasan A. Utility of dynamic susceptibility contrast MRI for differentiation between paragangliomas and meningiomas in the cerebellopontine angle and jugular foramen region. Clin Imaging 2023; 96:49-55. [PMID: 36801537 DOI: 10.1016/j.clinimag.2022.12.014] [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: 08/29/2022] [Revised: 12/21/2022] [Accepted: 12/27/2022] [Indexed: 01/15/2023]
Abstract
PURPOSE Differentiation of paragangliomas and meningiomas can be a challenge. This study aimed to assess the utility of dynamic susceptibility contrast perfusion MRI (DSC-MRI) to distinguish paragangliomas from meningiomas. METHODS This retrospective study included 40 patients with paragangliomas and meningiomas in the cerebellopontine angle and jugular foramen region between March 2015 and February 2022 in a single institution. Pretreatment DSC-MRI and conventional MRI were performed in all cases. Normalized relative cerebral blood volume (nrCBV), relative cerebral blood flow (nrCBF), relative mean transit time (nrMTT), and time to peak (nTTP) as well as conventional MRI features were compared between the 2 tumor types and between meningioma subtypes as appropriate. Receiver operating characteristic curve and multivariate logistic regression analysis were performed. RESULTS Twenty-eight meningiomas including 8 WHO grade II meningiomas (12 males, 16 females; median age 55 years) and 12 paragangliomas (5 males, 7 females; median age 35 years) were included in this study. Paragangliomas had a higher rate of cystic/necrotic changes (10/12 vs 10/28; P = 0.014), a higher rate of internal flow voids (9/12 vs 8/28; P = 0.013), higher nrCBV (median 9.78 vs 6.64; P = 0.04), and shorter nTTP (median 0.78 vs 1.06; P < 0.001) than meningiomas. There was no difference in conventional imaging features and DSC-MRI parameters between meningioma subtypes. nTTP was identified as the most significant parameter for the 2 tumor types in the multivariate logistic regression analysis (P = 0.009). CONCLUSIONS In this small retrospective study, DSC-MRI perfusion differences were observed between paragangliomas and meningiomas, but not between grade I and II meningiomas.
Collapse
Affiliation(s)
- Yoshiaki Ota
- Division of Neuroradiology, Department of Radiology, University of Michigan, 1500 E Medical Center Dr, UH B2, Ann Arbor, MI 48109, USA.
| | - Eric Liao
- Division of Neuroradiology, Department of Radiology, University of Michigan, 1500 E Medical Center Dr, UH B2, Ann Arbor, MI 48109, USA
| | - Raymond Zhao
- Division of Neuroradiology, Department of Radiology, University of Michigan, 1500 E Medical Center Dr, UH B2, Ann Arbor, MI 48109, USA
| | - Aristides A Capizzano
- Division of Neuroradiology, Department of Radiology, University of Michigan, 1500 E Medical Center Dr, UH B2, Ann Arbor, MI 48109, USA
| | - Akira Baba
- Division of Neuroradiology, Department of Radiology, University of Michigan, 1500 E Medical Center Dr, UH B2, Ann Arbor, MI 48109, USA
| | - Remy Lobo
- Division of Neuroradiology, Department of Radiology, University of Michigan, 1500 E Medical Center Dr, UH B2, Ann Arbor, MI 48109, USA
| | - Gaurang Shah
- Division of Neuroradiology, Department of Radiology, University of Michigan, 1500 E Medical Center Dr, UH B2, Ann Arbor, MI 48109, USA
| | - Ashok Srinivasan
- Division of Neuroradiology, Department of Radiology, University of Michigan, 1500 E Medical Center Dr, UH B2, Ann Arbor, MI 48109, USA
| |
Collapse
|
12
|
Witzmann K, Raschke F, Löck S, Wesemann T, Krause M, Linn J, Troost EGC. Unchanged perfusion in normal-appearing white and grey matter of glioma patients nine months after proton beam irradiation. Acta Oncol 2023; 62:141-149. [PMID: 36801809 DOI: 10.1080/0284186x.2023.2176254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Purpose: Radio(chemo)therapy is used as a standard treatment for glioma patients. The surrounding normal tissue is inevitably affected by the irradiation. The aim of this longitudinal study was to investigate perfusion alterations in the normal-appearing tissue after proton irradiation and assess the dose sensitivity of the normal tissue perfusion.Methods: In 14 glioma patients, a sub-cohort of a prospective clinical trial (NCT02824731), perfusion changes in normal-appearing white matter (WM), grey matter (GM) and subcortical GM structures, i.e. caudate nucleus, hippocampus, amygdala, putamen, pallidum and thalamus, were evaluated before treatment and at three-monthly intervals after proton beam irradiation. The relative cerebral blood volume (rCBV) was assessed with dynamic susceptibility contrast MRI and analysed as the percentage ratio between follow-up and baseline image (ΔrCBV). Radiation-induced alterations were evaluated using Wilcoxon signed rank test. Dose and time correlations were investigated with univariate and multivariate linear regression models.Results: No significant ΔrCBV changes were found in any normal-appearing WM and GM region after proton beam irradiation. A positive correlation with radiation dose was observed in the multivariate regression model applied to the combined ΔrCBV values of low (1-20 Gy), intermediate (21-40 Gy) and high (41-60 Gy) dose regions of GM (p < 0.001), while no time dependency was detected in any normal-appearing area.Conclusion: The perfusion in normal-appearing brain tissue remained unaltered after proton beam therapy. In further studies, a direct comparison with changes after photon therapy is recommended to confirm the different effect of proton therapy on the normal-appearing tissue.
Collapse
Affiliation(s)
- Katharina Witzmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany.,OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Felix Raschke
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany.,OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Steffen Löck
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and; Helmholtz Association / Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Tim Wesemann
- Institute of Neuroradiology, University Hospital Carl Gustav Carus and Medical Faculty of Technische Universität, Dresden, Germany
| | - Mechthild Krause
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany.,OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and; Helmholtz Association / Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Jennifer Linn
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and; Helmholtz Association / Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,Institute of Neuroradiology, University Hospital Carl Gustav Carus and Medical Faculty of Technische Universität, Dresden, Germany
| | - Esther G C Troost
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany.,OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and; Helmholtz Association / Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| |
Collapse
|
13
|
Baba A, Kurokawa R, Kurokawa M, Ota Y, Srinivasan A. Dynamic Contrast-Enhanced MRI Parameters and Normalized ADC Values Could Aid Differentiation of Skull Base Osteomyelitis from Nasopharyngeal Cancer. AJNR Am J Neuroradiol 2023; 44:74-78. [PMID: 36521963 PMCID: PMC9835913 DOI: 10.3174/ajnr.a7740] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/15/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND PURPOSE The skull base osteomyelitis sometimes can be difficult to distinguish from nasopharyngeal cancer. This study aimed to investigate the differences between skull base osteomyelitis and nasopharyngeal cancer using dynamic contrast-enhanced MR imaging and normalized ADC values. MATERIALS AND METHODS This study included 8 and 12 patients with skull base osteomyelitis and nasopharyngeal cancer, respectively, who underwent dynamic contrast-enhanced MR imaging and DWI before primary treatment. Quantitative dynamic contrast-enhanced MR imaging parameters and ADC values of the ROIs were analyzed. Normalized ADC parameters were calculated by dividing the ROIs of the lesion by that of the spinal cord. RESULTS The rate transfer constant between extravascular extracellular space and blood plasma per minute (Kep) was significantly lower in patients with skull base osteomyelitis than in those with nasopharyngeal cancer (median, 0.43 versus 0.57; P = .04). The optimal cutoff value of Kep was 0.48 (area under the curve, 0.78; 95% CI, 0.55-1). The normalized mean ADC was significantly higher in patients with skull base osteomyelitis than in those with nasopharyngeal cancer (median, 1.90 versus 0.87; P < .001). The cutoff value of normalized mean ADC was 1.55 (area under the curve, 0.96; 95% CI, 0.87-1). The area under the curve of the combination of dynamic contrast-enhanced MR imaging parameters (Kep and extravascular extracellular space volume per unit tissue volume) was 0.89 (95% CI, 0.73-1), and the area under the curve of the combination of dynamic contrast-enhanced MR imaging parameters and normalized mean ADC value was 0.98 (95% CI, 0.93-1). CONCLUSIONS Quantitative dynamic contrast-enhanced MR imaging parameters and normalized ADC values may be useful in differentiating skull base osteomyelitis and nasopharyngeal cancer. The combination of dynamic contrast-enhanced MR imaging parameters and normalized ADC values outperformed each measure in isolation.
Collapse
Affiliation(s)
- A Baba
- From the Division of Neuroradiology (A.B., R.K., M.K., Y.O., A.S.), Department of Radiology, University of Michigan, Ann Arbor, Michigan
- Department of Radiology (A.B.), The Jikei University School of Medicine, Tokyo, Japan
| | - R Kurokawa
- From the Division of Neuroradiology (A.B., R.K., M.K., Y.O., A.S.), Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - M Kurokawa
- From the Division of Neuroradiology (A.B., R.K., M.K., Y.O., A.S.), Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Y Ota
- From the Division of Neuroradiology (A.B., R.K., M.K., Y.O., A.S.), Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - A Srinivasan
- From the Division of Neuroradiology (A.B., R.K., M.K., Y.O., A.S.), Department of Radiology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
14
|
Kurokawa R, Kurokawa M, Baba A, Kim J, Capizzano A, Bapuraj J, Srinivasan A, Moritani T. Differentiation of pilocytic astrocytoma, medulloblastoma, and hemangioblastoma on diffusion-weighted and dynamic susceptibility contrast perfusion MRI. Medicine (Baltimore) 2022; 101:e31708. [PMID: 36343086 PMCID: PMC9646672 DOI: 10.1097/md.0000000000031708] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022] Open
Abstract
This study aimed to evaluate the diagnostic performance of dynamic susceptibility contrast (DSC) perfusion magnetic resonance imaging and apparent diffusion coefficient (ADC) for differentiating common posterior fossa tumors, pilocytic astrocytoma (PA), medulloblastoma (MB), and hemangioblastoma (HB). Between January 2016 and April 2022, we enrolled 23 (median age, 7 years [range, 2-26]; 12 female), 13 (10 years [1-24]; 3 female), and 12 (43 years [23-73]; 7 female) patients with PA, MB, and HB, respectively. Normalized relative cerebral blood volume and flow (nrCBV and nrCBF) and normalized mean ADC (nADCmean) were calculated from volume-of-interest and statistically compared. nADCmean was significantly higher in PA than in MB (PA: median, 2.2 [range, 1.59-2.65] vs MB: 0.93 [0.70-1.37], P < .001). nrCBF was significantly higher in HB than in PA and MB (PA: 1.10 [0.54-2.26] vs MB: 1.62 [0.93-3.16] vs HB: 7.83 [2.75-20.1], all P < .001). nrCBV was significantly different between all 3 tumor types (PA: 0.89 [0.34-2.28] vs MB: 1.69 [0.93-4.23] vs HB: 8.48 [4.59-16.3], P = .008 for PA vs MB; P < .001 for PA vs HB and MB vs HB). All tumors were successfully differentiated using an algorithmic approach with a threshold value of 4.58 for nrCBV and subsequent threshold value of 1.38 for nADCmean. DSC parameters and nADCmean were significantly different between PA, MB, and HB. An algorithmic approach combining nrCBV and nADCmean may be useful for differentiating these tumor types.
Collapse
Affiliation(s)
- Ryo Kurokawa
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Mariko Kurokawa
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Akira Baba
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - John Kim
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Aristides Capizzano
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jayapalli Bapuraj
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Ashok Srinivasan
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Toshio Moritani
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
15
|
Kurokawa R, Kurokawa M, Holmes A, Baba A, Bapuraj J, Capizzano A, Kim J, Srinivasan A, Moritani T. Skull metastases and osseous venous malformations: The role of diffusion-weighted and dynamic contrast-enhanced MRI. J Neuroimaging 2022; 32:1170-1176. [PMID: 35922879 DOI: 10.1111/jon.13034] [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: 07/01/2022] [Revised: 07/19/2022] [Accepted: 07/25/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Skull metastasis (SM) is a common secondary malignancy. We evaluated the diagnostic performance of diffusion-weighted imaging (DWI) and dynamic contrast-enhanced magnetic resonance imaging (MRI) in differentiating SM from osseous venous malformations and SM of various origins. METHODS This study included 31 patients with SM (median age, 64 years; range, 41-87 years; 29 women; 24 and 7 patients with breast and non-small cell lung cancer, respectively) and 16 with osseous venous malformations (median age, 68 years; range, 20-81 years; 10 women) who underwent both DWI and dynamic contrast-enhanced MRI between January 2015 and October 2021. Normalized mean apparent diffusion coefficients (ADCs) and dynamic contrast-enhanced MRI parameters were compared between SM and osseous venous malformations, and between breast cancer and non-small cell lung cancer. Multivariate stepwise logistic regression analyses were performed to identify statistically significant parameters. RESULTS Plasma volume and time-to-maximum enhancement were the most statistically significant parameters for differentiating SM from osseous venous malformations, with an area under the receiver operating characteristic curve of 0.962. The normalized mean ADC and peak enhancement values were the most statistically significant parameters for differentiating breast cancer from non-small cell lung cancer, with an area under the curve of 0.924. CONCLUSIONS Our results highlight the efficacious diagnostic performance of DWI and dynamic contrast-enhanced MRI in distinguishing SM from osseous venous malformations and differentiating SM of various origins.
Collapse
Affiliation(s)
- Ryo Kurokawa
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Mariko Kurokawa
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Adam Holmes
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Akira Baba
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jayapalli Bapuraj
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Aristides Capizzano
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - John Kim
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Ashok Srinivasan
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Toshio Moritani
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
16
|
Dong W, Volk A, Djaroum M, Girot C, Balleyguier C, Lebon V, Garcia G, Ammari S, Temam S, Gorphe P, Wei L, Pitre-Champagnat S, Lassau N, Bidault F. Influence of Different Measurement Methods of Arterial Input Function on Quantitative Dynamic Contrast-Enhanced MRI Parameters in Head and Neck Cancer. J Magn Reson Imaging 2022. [PMID: 36269053 DOI: 10.1002/jmri.28486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Head and neck cancer (HNC) is the sixth most prevalent cancer worldwide. Dynamic contrast-enhanced MRI (DCE-MRI) helps in diagnosis and prognosis. Quantitative DCE-MRI requires an arterial input function (AIF), which affects the values of pharmacokinetic parameters (PKP). PURPOSE To evaluate influence of four individual AIF measurement methods on quantitative DCE-MRI parameters values (Ktrans , ve , kep , and vp ), for HNC and muscle. STUDY TYPE Prospective. POPULATION A total of 34 HNC patients (23 males, 11 females, age range 24-91) FIELD STRENGTH/SEQUENCE: A 3 T; 3D SPGR gradient echo sequence with partial saturation of inflowing spins. ASSESSMENT Four AIF methods were applied: automatic AIF (AIFa) with up to 50 voxels selected from the whole FOV, manual AIF (AIFm) with four voxels selected from the internal carotid artery, both conditions without (Mc-) or with (Mc+) motion correction. Comparison endpoints were peak AIF values, PKP values in tumor and muscle, and tumor/muscle PKP ratios. STATISTICAL TESTS Nonparametric Friedman test for multiple comparisons. Nonparametric Wilcoxon test, without and with Benjamini Hochberg correction, for pairwise comparison of AIF peak values and PKP values for tumor, muscle and tumor/muscle ratio, P value ≤ 0.05 was considered statistically significant. RESULTS Peak AIF values differed significantly for all AIF methods, with mean AIFmMc+ peaks being up to 66.4% higher than those for AIFaMc+. Almost all PKP values were significantly higher for AIFa in both, tumor and muscle, up to 76% for mean Ktrans values. Motion correction effect was smaller. Considering tumor/muscle parameter ratios, most differences were not significant (0.068 ≤ Wilcoxon P value ≤ 0.8). DATA CONCLUSION We observed important differences in PKP values when using either AIFa or AIFm, consequently choice of a standardized AIF method is mandatory for DCE-MRI on HNC. From the study findings, AIFm and inflow compensation are recommended. The use of the tumor/muscle PKP ratio should be of interest for multicenter studies. EVIDENCE LEVEL 2 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Wanxin Dong
- Multimodal Biomedical Imaging Laboratory (BioMaps), Paris-Saclay University, Inserm (UMR1281), CNRS (UMR9011), CEA, France
| | - Andreas Volk
- Multimodal Biomedical Imaging Laboratory (BioMaps), Paris-Saclay University, Inserm (UMR1281), CNRS (UMR9011), CEA, France
| | - Meriem Djaroum
- Multimodal Biomedical Imaging Laboratory (BioMaps), Paris-Saclay University, Inserm (UMR1281), CNRS (UMR9011), CEA, France
| | - Charly Girot
- Multimodal Biomedical Imaging Laboratory (BioMaps), Paris-Saclay University, Inserm (UMR1281), CNRS (UMR9011), CEA, France
| | - Corinne Balleyguier
- Multimodal Biomedical Imaging Laboratory (BioMaps), Paris-Saclay University, Inserm (UMR1281), CNRS (UMR9011), CEA, France.,Department of Medical Imaging, Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France
| | - Vincent Lebon
- Multimodal Biomedical Imaging Laboratory (BioMaps), Paris-Saclay University, Inserm (UMR1281), CNRS (UMR9011), CEA, France
| | - Gabriel Garcia
- Department of Medical Imaging, Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France
| | - Samy Ammari
- Multimodal Biomedical Imaging Laboratory (BioMaps), Paris-Saclay University, Inserm (UMR1281), CNRS (UMR9011), CEA, France.,Department of Medical Imaging, Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France
| | - Stéphane Temam
- Department of Head and Neck Oncology, Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France
| | - Philippe Gorphe
- Department of Head and Neck Oncology, Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France
| | - Lecong Wei
- Multimodal Biomedical Imaging Laboratory (BioMaps), Paris-Saclay University, Inserm (UMR1281), CNRS (UMR9011), CEA, France
| | - Stéphanie Pitre-Champagnat
- Multimodal Biomedical Imaging Laboratory (BioMaps), Paris-Saclay University, Inserm (UMR1281), CNRS (UMR9011), CEA, France
| | - Nathalie Lassau
- Multimodal Biomedical Imaging Laboratory (BioMaps), Paris-Saclay University, Inserm (UMR1281), CNRS (UMR9011), CEA, France.,Department of Medical Imaging, Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France
| | - François Bidault
- Multimodal Biomedical Imaging Laboratory (BioMaps), Paris-Saclay University, Inserm (UMR1281), CNRS (UMR9011), CEA, France.,Department of Medical Imaging, Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France
| |
Collapse
|
17
|
Juttukonda MR, Stephens KA, Yen YF, Howard CM, Polimeni JR, Rosen BR, Salat DH. Oxygen extraction efficiency and white matter lesion burden in older adults exhibiting radiological evidence of capillary shunting. J Cereb Blood Flow Metab 2022; 42:1933-1943. [PMID: 35673981 PMCID: PMC9536117 DOI: 10.1177/0271678x221105986] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/19/2022] [Accepted: 05/14/2022] [Indexed: 01/18/2023]
Abstract
White matter lesions (WML) have been linked to cognitive decline in aging as well as in Alzheimer's disease. While hypoperfusion is frequently considered a cause of WMLs due to the resulting reduction in oxygen availability to brain tissue, such reductions could also be caused by impaired oxygen exchange. Here, we tested the hypothesis that venous hyperintense signal (VHS) in arterial spin labeling (ASL) magnetic resonance imaging (MRI) may represent a marker of impaired oxygen extraction in aging older adults. In participants aged 60-80 years (n = 30), we measured cerebral blood flow and VHS with arterial spin labeling, maximum oxygen extraction fraction (OEFmax) with dynamic susceptibility contrast, and WML volume with T1-weighted MRI. We found a significant interaction between OEFmax and VHS presence on WML volume (p = 0.02), where lower OEFmax was associated with higher WML volume in participants with VHS, and higher OEFmax was associated with higher WML volume in participants without VHS. These results indicate that VHS in perfusion-weighted ASL data may represent a distinct cerebrovascular aging pattern involving oxygen extraction inefficiency as well as hypoperfusion.
Collapse
Affiliation(s)
- Meher R Juttukonda
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Kimberly A Stephens
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Yi-Fen Yen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Casey M Howard
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Jonathan R Polimeni
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
- Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bruce R Rosen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
- Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David H Salat
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
- Neuroimaging Research for Veterans Center, VA Boston Healthcare System, Boston, MA, USA
| |
Collapse
|
18
|
Kurokawa R, Baba A, Kurokawa M, Capizzano A, Ota Y, Kim J, Srinivasan A, Moritani T. Perfusion and diffusion-weighted imaging parameters: Comparison between pre- and postbiopsy MRI for high-grade glioma. Medicine (Baltimore) 2022; 101:e30183. [PMID: 36107564 PMCID: PMC9439799 DOI: 10.1097/md.0000000000030183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
We aimed to evaluate the differences in dynamic susceptibility contrast (DSC)- magnetic resonance imaging (MRI) and diffusion-weighted imaging (DWI) parameters between the pre- and postbiopsy MRI obtained before treatment in patients with diffuse midline glioma, H3K27-altered. The data of 25 patients with pathologically proven diffuse midline glioma, H3K27-altered, were extracted from our hospital's database between January 2017 and August 2021. Twenty (median age, 13 years; range, 3-52 years; 12 women) and 8 (13.5 years; 5-68 years; 1 woman) patients underwent preoperative DSC-MRI and DWI before and after biopsy, respectively. The normalized corrected relative cerebral blood volume (ncrCBV), normalized relative cerebral blood flow (nrCBF), and normalized maximum, mean, and minimum apparent diffusion coefficient (ADC) were calculated using the volumes-of-interest of the tumor and normal-appearing reference region. The macroscopic postbiopsy changes (i.e., biopsy tract, tissue defect, and hemorrhage) were meticulously excluded from the postbiopsy measurements. The DSC-MRI and DWI parameters of the pre- and postbiopsy groups were compared using the Mann-Whitney U test. The ncrCBV was significantly lower in the postbiopsy group than in the prebiopsy group [prebiopsy group: median 1.293 (range, 0.513 to 2.547) versus postbiopsy group: 0.877 (0.748 to 1.205), P = .016]. No significant difference was observed in the nrCBF and normalized ADC values, although the median nrCBF was lower in the postbiopsy group. The DSC-MRI parameters differed between the pre- and postbiopsy MRI obtained pretreatment, although the macroscopic postbiopsy changes were carefully excluded from the analysis. The results emphasize the potential danger of integrating and analyzing DSC-MRI parameters derived from pre- and postbiopsy MRI.
Collapse
Affiliation(s)
- Ryo Kurokawa
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, MI
- *Correspondence: Ryo Kurokawa, Division of Neuroradiology, Department of Radiology, University of Michigan, 1500 E Medical Center Dr, UH B2, Ann Arbor, MI 48109 (e-mail: )
| | - Akira Baba
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, MI
| | - Mariko Kurokawa
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, MI
| | - Aristides Capizzano
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, MI
| | - Yoshiaki Ota
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, MI
| | - John Kim
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, MI
| | - Ashok Srinivasan
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, MI
| | - Toshio Moritani
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
19
|
Zhou X, Nan Y, Ju J, Zhou J, Xiao H, Wang S. Comparison of Two Software Packages for Perfusion Imaging: Ischemic Core and Penumbra Estimation and Patient Triage in Acute Ischemic Stroke. Cells 2022; 11:2547. [PMID: 36010624 PMCID: PMC9406974 DOI: 10.3390/cells11162547] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/20/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
Purpose: Automated postprocessing packages have been developed for managing acute ischemic stroke (AIS). These packages identify ischemic core and penumbra using either computed tomographic perfusion imaging (CTP) data or magnetic resonance imaging (MRI) data. Measurements of abnormal tissues and treatment decisions derived from different vendors can vary. The purpose of this study is to investigate the agreement of volumetric and decision-making outcomes derived from two software packages. Methods: A total of 594 AIS patients (174 underwent CTP and 420 underwent MRI) were included. Imaging data were accordingly postprocessed by two software packages: RAPID and RealNow. Volumetric outputs were compared between packages by performing intraclass correlation coefficient (ICC), Wilcoxon paired test and Bland-Altman analysis. Concordance of selecting patients eligible for mechanical thrombectomy (MT) was assessed based on neuroimaging criteria proposed in DEFUSE3. Results: In the group with CTP data, mean ischemic core volume (ICV)/penumbral volume (PV) was 14.9/81.1 mL via RAPID and 12.6/83.2 mL via RealNow. Meanwhile, in the MRI group, mean ICV/PV were 52.4/68.4 mL and 48.9/61.6 mL via RAPID and RealNow, respectively. Reliability, which was measured by ICC of ICV and PV in CTP and MRI groups, ranged from 0.87 to 0.99. The bias remained small between measurements (CTP ICV: 0.89 mL, CTP PV: -2 mL, MRI ICV: 3.5 mL and MRI PV: 6.8 mL). In comparison with CTP ICV with follow-up DWI, the ICC was 0.92 and 0.94 for RAPID and Realnow, respectively. The bias remained small between CTP ICV and follow-up DWI measurements (Rapid: -4.65 mL, RealNow: -3.65 mL). Wilcoxon paired test showed no significant difference between measurements. The results of patient triage were concordant in 159/174 cases (91%, ICC: 0.90) for CTP and 400/420 cases (95%, ICC: 0.93) for MRI. Conclusion: The CTP ICV derived from RealNow was more accurate than RAPID. The similarity in volumetric measurement between packages did not necessarily relate to equivalent patient triage. In this study, RealNow showed excellent agreement with RAPID in measuring ICV and PV as well as patient triage.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Radiology, Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Rd., Shanghai 200065, China
| | - Yashi Nan
- YIWEI Medical Technology Co., Ltd., Room 1001, MAI KE LONG Building, Shenzhen 518000, China
| | - Jieyang Ju
- The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjiayuan Rd., Nanjing 210011, China
| | - Jingyu Zhou
- YIWEI Medical Technology Co., Ltd., Room 1001, MAI KE LONG Building, Shenzhen 518000, China
| | - Huanhui Xiao
- YIWEI Medical Technology Co., Ltd., Room 1001, MAI KE LONG Building, Shenzhen 518000, China
| | - Silun Wang
- YIWEI Medical Technology Co., Ltd., Room 1001, MAI KE LONG Building, Shenzhen 518000, China
| |
Collapse
|
20
|
Baba A, Kurokawa R, Rawie E, Kurokawa M, Ota Y, Srinivasan A. Normalized Parameters of Dynamic Contrast-Enhanced Perfusion MRI and DWI-ADC for Differentiation between Posttreatment Changes and Recurrence in Head and Neck Cancer. AJNR Am J Neuroradiol 2022; 43:1184-1189. [PMID: 35835592 PMCID: PMC9575415 DOI: 10.3174/ajnr.a7567] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 05/22/2022] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND PURPOSE Differentiating recurrence from benign posttreatment changes has clinical importance in the imaging follow-up of head and neck cancer. This study aimed to investigate the utility of normalized dynamic contrast-enhanced MR imaging and ADC for their differentiation. MATERIALS AND METHODS This study included 51 patients with a history of head and neck cancer who underwent follow-up dynamic contrast-enhanced MR imaging with DWI-ADC, of whom 25 had recurrences and 26 had benign posttreatment changes. Quantitative and semiquantitative dynamic contrast-enhanced MR imaging parameters and ADC of the ROI and reference region were analyzed. Normalized dynamic contrast-enhanced MR imaging parameters and normalized DWI-ADC parameters were calculated by dividing the ROI by the reference region. RESULTS Normalized plasma volume, volume transfer constant between extravascular extracellular space and blood plasma per minute (K trans), area under the curve, and wash-in were significantly higher in patients with recurrence than in those with benign posttreatment change (P = .003 to <.001). The normalized mean ADC was significantly lower in patients with recurrence than in those with benign posttreatment change (P < .001). The area under the receiver operating characteristic curve of the combination of normalized dynamic contrast-enhanced MR imaging parameters with significance (normalized plasma volume, normalized extravascular extracellular space volume per unit tissue volume, normalized K trans, normalized area under the curve, and normalized wash-in) and normalized mean ADC was 0.97 (95% CI, 0.93-1). CONCLUSIONS Normalized dynamic contrast-enhanced MR imaging parameters, normalized mean ADC, and their combination were effective in differentiating recurrence and benign posttreatment changes in head and neck cancer.
Collapse
Affiliation(s)
- A Baba
- From the Division of Neuroradiology (A.B., R.K., M.K., Y.O., A.S.), Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - R Kurokawa
- From the Division of Neuroradiology (A.B., R.K., M.K., Y.O., A.S.), Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - E Rawie
- Department of Radiology (E.R.), Brooke Army Medical Center, San Antonio, Texas
| | - M Kurokawa
- From the Division of Neuroradiology (A.B., R.K., M.K., Y.O., A.S.), Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Y Ota
- From the Division of Neuroradiology (A.B., R.K., M.K., Y.O., A.S.), Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - A Srinivasan
- From the Division of Neuroradiology (A.B., R.K., M.K., Y.O., A.S.), Department of Radiology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
21
|
Starck L, Skeie BS, Moen G, Grüner R. Dynamic Susceptibility Contrast MRI May Contribute in Prediction of Stereotactic Radiosurgery Outcome in Brain Metastases. Neurooncol Adv 2022; 4:vdac070. [PMID: 35673606 PMCID: PMC9167634 DOI: 10.1093/noajnl/vdac070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background Following stereotactic radiosurgery (SRS), predicting treatment response is not possible at an early stage using structural imaging alone. Hence, the current study aims at investigating whether dynamic susceptibility contrast (DSC)-MRI estimated prior to SRS can provide predictive biomarkers in response to SRS treatment and characterize vascular characteristics of pseudo-progression. Methods In this retrospective study, perfusion-weighted DSC-MRI image data acquired with a temporal resolution of 1.45 seconds were collected from 41 patients suffering from brain metastases. Outcome was defined based on lesion volume changes in time (determined on structural images) or death. Motion correction and manual lesion delineation were performed prior to semi-automated, voxel-wise perfusion analysis. Statistical testing was performed using linear regression and a significance threshold at P = .05. Age, sex, primary cancers (pulmonary cancer and melanoma), lesion volume, and dichotomized survival time were added as covariates in the linear regression models (ANOVA). Results Relative cerebral blood volume (rCBV) and relative cerebral blood flow (rCBF) were found to be significantly lower prior to SRS treatment in patients with increasing lesion volume or early death post-SRS (P ≤ .01). Conclusion Unfavorable treatment outcome may be linked to low perfusion prior to SRS. Pseudo-progression may be preceded by a transient rCBF increase post-SRS. However, results should be verified in different or larger patient material.
Collapse
Affiliation(s)
- Lea Starck
- Department of Physics and Technology, University of Bergen, Norway
- Mohn Medical Imaging and Visualization Centre, Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | | | - Gunnar Moen
- Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Renate Grüner
- Department of Physics and Technology, University of Bergen, Norway
- Mohn Medical Imaging and Visualization Centre, Department of Radiology, Haukeland University Hospital, Bergen, Norway
- Department of Radiology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
22
|
Bae J, Huang Z, Knoll F, Geras K, Sood TP, Feng L, Heacock L, Moy L, Kim SG. Estimation of the capillary level input function for dynamic contrast-enhanced MRI of the breast using a deep learning approach. Magn Reson Med 2022; 87:2536-2550. [PMID: 35001423 PMCID: PMC8852816 DOI: 10.1002/mrm.29148] [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: 01/24/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 11/06/2022]
Abstract
PURPOSE To develop a deep learning approach to estimate the local capillary-level input function (CIF) for pharmacokinetic model analysis of DCE-MRI. METHODS A deep convolutional network was trained with numerically simulated data to estimate the CIF. The trained network was tested using simulated lesion data and used to estimate voxel-wise CIF for pharmacokinetic model analysis of breast DCE-MRI data using an abbreviated protocol from women with malignant (n = 25) and benign (n = 28) lesions. The estimated parameters were used to build a logistic regression model to detect the malignancy. RESULT The pharmacokinetic parameters estimated using the network-predicted CIF from our breast DCE data showed significant differences between the malignant and benign groups for all parameters. Testing the diagnostic performance with the estimated parameters, the conventional approach with arterial input function (AIF) showed an area under the curve (AUC) between 0.76 and 0.87, and the proposed approach with CIF demonstrated similar performance with an AUC between 0.79 and 0.81. CONCLUSION This study shows the feasibility of estimating voxel-wise CIF using a deep neural network. The proposed approach could eliminate the need to measure AIF manually without compromising the diagnostic performance to detect the malignancy in the clinical setting.
Collapse
Affiliation(s)
- Jonghyun Bae
- Vilcek Institute of Graduate Biomedical Science, New York University School of Medicine
- Center for Biomedical Imaging, Radiology, New York University School of Medicine
- Center for Advanced Imaging Innovation and Research, Radiology, New York University School of Medicine
- Department of Radiology, Weill Cornell Medical College
| | - Zhengnan Huang
- Vilcek Institute of Graduate Biomedical Science, New York University School of Medicine
- Center for Biomedical Imaging, Radiology, New York University School of Medicine
- Center for Advanced Imaging Innovation and Research, Radiology, New York University School of Medicine
| | - Florian Knoll
- Center for Biomedical Imaging, Radiology, New York University School of Medicine
- Center for Advanced Imaging Innovation and Research, Radiology, New York University School of Medicine
| | - Krzysztof Geras
- Center for Biomedical Imaging, Radiology, New York University School of Medicine
- Center for Advanced Imaging Innovation and Research, Radiology, New York University School of Medicine
- Center for Data Science, New York University
| | - Terlika Pandit Sood
- Center for Biomedical Imaging, Radiology, New York University School of Medicine
- Center for Advanced Imaging Innovation and Research, Radiology, New York University School of Medicine
| | - Li Feng
- Biomedical Engineering and Imaging Institute and Department of Radiology, Icahn School of Medicine at Mount Sinai
| | - Laura Heacock
- Center for Biomedical Imaging, Radiology, New York University School of Medicine
- Center for Advanced Imaging Innovation and Research, Radiology, New York University School of Medicine
| | - Linda Moy
- Vilcek Institute of Graduate Biomedical Science, New York University School of Medicine
- Center for Biomedical Imaging, Radiology, New York University School of Medicine
- Center for Advanced Imaging Innovation and Research, Radiology, New York University School of Medicine
| | | |
Collapse
|
23
|
Ota Y, Moore AG, Spector ME, Casper K, Stucken C, Malloy K, Lobo R, Baba A, Srinivasan A. Prediction of Wound Failure in Patients with Head and Neck Cancer Treated with Free Flap Reconstruction: Utility of CT Perfusion and MR Perfusion in the Early Postoperative Period. AJNR Am J Neuroradiol 2022; 43:585-591. [PMID: 35361578 PMCID: PMC8993192 DOI: 10.3174/ajnr.a7458] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/08/2022] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND PURPOSE Free flap reconstruction in patients with head and neck cancer carries a risk of postoperative complications, and radiologic predictive factors have been limited. The aim of this study was to assess the factors that predict free flap reconstruction failure using CT and MR perfusion. MATERIALS AND METHODS This single-center prospective study included 24 patients (mean age, 62.7 [SD, 9.0] years; 16 men) who had free flap reconstruction from January 2016 to May 2018. CT perfusion and dynamic contrast-enhanced MR imaging with conventional CT and MR imaging were performed between 2 and 4 days after the free flap surgery, and the wound assessments within 14 days after the surgery were conducted by the surgical team. The parameters of CT perfusion and dynamic contrast-enhanced MR imaging with conventional imaging findings and patient demographics were compared between the patients with successful free flap reconstruction and those with wound failure as appropriate. P < .05 was considered significant. RESULTS There were 19 patients with successful free flap reconstruction and no wound complications (mean age, 63.9 [SD, 9.5] years; 14 men), while 5 patients had wound failure (mean age, 58.0 [SD, 5.7] years; 2 men). Blood flow, blood volume, MTT, and time maximum intensity projection (P = .007, .007, .015, and .004, respectively) in CT perfusion, and fractional plasma volume, volume transfer constant, peak enhancement, and time to maximum enhancement (P = .006, .039, .004, and .04, respectively) in dynamic contrast-enhanced MR imaging were significantly different between the 2 groups. CONCLUSIONS CT perfusion and dynamic contrast-enhanced MR imaging are both promising imaging techniques to predict wound complications after head and neck free flap reconstruction.
Collapse
Affiliation(s)
- Y Ota
- From the Division of Neuroradiology (Y.O., R.L., A.B., A.S.)
| | - A G Moore
- Department of Radiology (A.G.M.), Western Michigan University, Kalamazoo, Michigan
| | - M E Spector
- Department of Radiology, and Department of Otolaryngology (M.E.S., K.C., C.S., K.M.), University of Michigan, Ann Arbor, Michigan
| | - K Casper
- Department of Radiology, and Department of Otolaryngology (M.E.S., K.C., C.S., K.M.), University of Michigan, Ann Arbor, Michigan
| | - C Stucken
- Department of Radiology, and Department of Otolaryngology (M.E.S., K.C., C.S., K.M.), University of Michigan, Ann Arbor, Michigan
| | - K Malloy
- Department of Radiology, and Department of Otolaryngology (M.E.S., K.C., C.S., K.M.), University of Michigan, Ann Arbor, Michigan
| | - R Lobo
- From the Division of Neuroradiology (Y.O., R.L., A.B., A.S.)
| | - A Baba
- From the Division of Neuroradiology (Y.O., R.L., A.B., A.S.)
| | - A Srinivasan
- From the Division of Neuroradiology (Y.O., R.L., A.B., A.S.)
| |
Collapse
|
24
|
Bhaduri S, Lesbats C, Sharkey J, Kelly CL, Mukherjee S, Taylor A, Delikatny EJ, Kim SG, Poptani H. Assessing Tumour Haemodynamic Heterogeneity and Response to Choline Kinase Inhibition Using Clustered Dynamic Contrast Enhanced MRI Parameters in Rodent Models of Glioblastoma. Cancers (Basel) 2022; 14:cancers14051223. [PMID: 35267531 PMCID: PMC8909848 DOI: 10.3390/cancers14051223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/16/2022] [Accepted: 02/23/2022] [Indexed: 12/04/2022] Open
Abstract
To investigate the utility of DCE-MRI derived pharmacokinetic parameters in evaluating tumour haemodynamic heterogeneity and treatment response in rodent models of glioblastoma, imaging was performed on intracranial F98 and GL261 glioblastoma bearing rodents. Clustering of the DCE-MRI-based parametric maps (using Tofts, extended Tofts, shutter speed, two-compartment, and the second generation shutter speed models) was performed using a hierarchical clustering algorithm, resulting in areas with poor fit (reflecting necrosis), low, medium, and high valued pixels representing parameters Ktrans, ve, Kep, vp, τi and Fp. There was a significant increase in the number of necrotic pixels with increasing tumour volume and a significant correlation between ve and tumour volume suggesting increased extracellular volume in larger tumours. In terms of therapeutic response in F98 rat GBMs, a sustained decrease in permeability and perfusion and a reduced cell density was observed during treatment with JAS239 based on Ktrans, Fp and ve as compared to control animals. No significant differences in these parameters were found for the GL261 tumour, indicating that this model may be less sensitive to JAS239 treatment regarding changes in vascular parameters. This study demonstrates that region-based clustered pharmacokinetic parameters derived from DCE-MRI may be useful in assessing tumour haemodynamic heterogeneity with the potential for assessing therapeutic response.
Collapse
Affiliation(s)
- Sourav Bhaduri
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
| | - Clémentine Lesbats
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SM2 5NG, UK
| | - Jack Sharkey
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
| | - Claire Louise Kelly
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
| | - Soham Mukherjee
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
| | - Arthur Taylor
- Department of Molecular Physiology & Cell Signalling, University of Liverpool, Liverpool L69 3BX, UK;
| | - Edward J. Delikatny
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Sungheon G. Kim
- Department of Radiology, Weill Cornell Medical College, New York, NY 10021, USA;
| | - Harish Poptani
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
- Correspondence:
| |
Collapse
|
25
|
Rotkopf LT, Zhang KS, Tavakoli AA, Bonekamp D, Ziener CH, Schlemmer HP. Quantitative Analysis of DCE and DSC-MRI: From Kinetic Modeling to Deep Learning. ROFO-FORTSCHR RONTG 2022; 194:975-982. [PMID: 35211930 DOI: 10.1055/a-1762-5854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Perfusion MRI is a well-established imaging modality with a multitude of applications in oncological and cardiovascular imaging. Clinically used processing methods, while stable and robust, have remained largely unchanged in recent years. Despite promising results from novel methods, their relatively minimal improvement compared to established methods did not generally warrant significant changes to clinical perfusion processing. RESULTS AND CONCLUSION Machine learning in general and deep learning in particular, which are currently revolutionizing computer-aided diagnosis, may carry the potential to change this situation and truly capture the potential of perfusion imaging. Recent advances in the training of recurrent neural networks make it possible to predict and classify time series data with high accuracy. Combining physics-based tissue models and deep learning, using either physics-informed neural networks or universal differential equations, simplifies the training process and increases the interpretability of the resulting models. Due to their versatility, these methods will potentially be useful in bridging the gap between microvascular architecture and perfusion parameters, akin to MR fingerprinting in structural MR imaging. Still, further research is urgently needed before these methods may be used in clinical practice. KEY POINTS · Machine learning offers promising methods for processing of perfusion data.. · Recurrent neural networks can classify time series with high accuracy.. · Data augmentation is essentially especially when using small datasets.. CITATION FORMAT · Rotkopf LT, Zhang KS, Tavakoli AA et al. Quantitative Analysis of DCE and DSC-MRI: From Kinetic Modeling to Deep Learning. Fortschr Röntgenstr 2022; DOI: 10.1055/a-1762-5854.
Collapse
Affiliation(s)
- Lukas T Rotkopf
- Department of Radiology, German Cancer Research Centre, Heidelberg, Germany
| | - Kevin Sun Zhang
- Department of Radiology, German Cancer Research Centre, Heidelberg, Germany
| | | | - David Bonekamp
- Department of Radiology, German Cancer Research Centre, Heidelberg, Germany
| | | | | |
Collapse
|
26
|
Şahin S, Ertekin E, Şahin T, Özsunar Y. Evaluation of normal-appearing white matter with perfusion and diffusion MRI in patients with treated glioblastoma. MAGMA (NEW YORK, N.Y.) 2022; 35:153-162. [PMID: 34951690 DOI: 10.1007/s10334-021-00990-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 12/10/2021] [Accepted: 12/12/2021] [Indexed: 06/14/2023]
Abstract
OBJECTIVE We tried to reveal how the normal appearing white matter (NAWM) was affected in patients with glioblastoma treated with chemo-radiotherapy (CRT) in the period following the treatment, by multiparametric MRI. MATERIALS AND METHODS 43 multiparametric MRI examinations of 17 patients with glioblastoma treated with CRT were examined. A total of six different series or maps were analyzed in the examinations: Apparent Diffusion Coefficient (ADC) and Fractional Anisotropy (FA) maps, Gradient Echo (GRE) sequence, Dynamic susceptibility contrast (DSC) and Arterial spin labeling (ASL) perfusion sequences. Each sequence in each examination was examined in detail with 14 Region of Interest (ROI) measurements. The obtained values were proportioned to the contralateral NAWM values and the results were recorded as normalized values. Time dependent changes of normalized values were statistically analyzed. RESULTS The most prominent changes in follow-up imaging occurred in the perilesional region. In perilesional NAWM, we found a decrease in normalized FA (nFA), rCBV (nrCBV), rCBF (nrCBF), ASL (nASL)values (p < 0.005) in the first 3 months after treatment, followed by a plateau and an increase approaching pretreatment values, although it did not reach. Similar but milder findings were present in other NAWM areas. In perilesional NAWM, nrCBV values were found to be positively high correlated with nrCBF and nASL, and negatively high correlated with nADC values (r: 0.963, 0.736, - 0.973, respectively). We also found high correlations between the mean values of nrCBV, nrCBF, nASL in other NAWM areas (r: 0.891, 0.864, respectively). DISCUSSION We showed that both DSC and ASL perfusion values decreased correlatively in the first 3 months and showed a plateau after 1 year in patients with glioblastoma treated with CRT, unlike the literature. Although it was not as evident as perfusion MRI, it was observed that the ADC values also showed a plateau pattern following the increase in the first 3 months. Further studies are needed to explain late pathophysiological changes. Because of the high correlation, our results support ASL perfusion instead of contrast enhanced perfusion methods.
Collapse
Affiliation(s)
- Sinan Şahin
- Department of Radiology, Adnan Menderes University, Aydın, Turkey
| | - Ersen Ertekin
- Department of Radiology, Adnan Menderes University, Aydın, Turkey.
| | - Tuna Şahin
- Department of Radiology, Adnan Menderes University, Aydın, Turkey
| | - Yelda Özsunar
- Department of Radiology, Adnan Menderes University, Aydın, Turkey
| |
Collapse
|
27
|
Bal SS, Chen K, Yang FPG, Peng G. Arterial Input Function segmentation based on a contour geodesic model for tissue at risk identification in Ischemic Stroke. Med Phys 2022; 49:2475-2485. [DOI: 10.1002/mp.15508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 11/10/2022] Open
Affiliation(s)
- Sukhdeep Singh Bal
- Department of Mathematical sciences University of Liverpool Liverpool UK
- Center for Cognition and Mind Sciences National Tsing Hua University Taiwan
- International Intercollegiate Ph.D. Programme National Tsing Hua University Taiwan
| | - Ke Chen
- Department of Mathematical sciences University of Liverpool Liverpool Merseyside UK
| | - Fan Pei Gloria Yang
- Center for Cognition and Mind Sciences National Tsing Hua University Taiwan
- Department of Foreign Languages and Literature National Tsing Hua University Taiwan
- Department of Radiology Graduate School of Dentistry Osaka University Japan
- No. 101, Section 2, Guangfu Road, East District Hsinchu City 300 Taiwan
| | - Giia‐Sheun Peng
- Department of Neurology Tri‐Service General Hospital National Defense Medical Center Taipei Taiwan
- Division of Neurology Department of Internal Medicine Taipei Veterans General Hospital Hsinchu Branch Hsinchu County Taiwan
| |
Collapse
|
28
|
Kurokawa R, Kurokawa M, Baba A, Ota Y, Kim J, Capizzano A, Srinivasan A, Moritani T. Dynamic susceptibility contrast-MRI parameters, ADC values, and the T2-FLAIR mismatch sign are useful to differentiate between H3-mutant and H3-wild-type high-grade midline glioma. Eur Radiol 2022; 32:3672-3682. [PMID: 35022811 DOI: 10.1007/s00330-021-08476-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/25/2021] [Accepted: 11/21/2021] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Diffuse midline gliomas, H3K27-altered (DMG-A), are malignant gliomas with an unfavorable prognosis. Knowledge of dynamic susceptibility contrast (DSC) MRI findings and imaging differences with high-grade midline glioma without H3K27 alteration (DMG-W) has been limited. We compared the DSC, ADC, and conventional MRI findings between DMG-A and DMG-W. METHODS In this single institutional retrospective study, the electronic database of our hospital between June 2015 and May 2021 was searched. Twenty and 17 patients with DMG-A (median, 13 years; range, 3-52 years; 11 females) and DMG-W (median, 40 years; 7-73 years; 9 females), respectively, were found. Normalized relative cerebral blood flow (nrCBF) and normalized corrected relative cerebral blood volume (ncrCBV); normalized maximum, mean, and minimum ADC values; and the prevalence of T2-FLAIR mismatch sign were compared between the two groups using Mann-Whitney U tests and Fisher's exact test. RESULTS The nrCBF and ncrCBV were significantly lower in DMG-A compared with DMG-W (nrCBF: median 0.88 [range, 0.19-2.67] vs. 1.47 [range, 0.57-4.90] (p < 0.001); ncrCBV: 1.17 [0.20-2.67] vs. 1.56 [0.60-4.03] (p = 0.008)). Normalized maximum ADC (nADCmax) was significantly higher in DMG-A (median 2.37 [1.25-3.98] vs. 1.95 [1.23-2.77], p = 0.02). T2-FLAIR mismatch sign was significantly more common in DMG-A (11/20 (55.0%) vs. 1/17 (5.9%), p = 0.0017). When at least two of nrCBF < 1.11, nADCmax ≥ 2.48, and T2-FLAIR mismatch sign were positive, the diagnostic performance was the highest with accuracy of 0.81. CONCLUSION DSC-MRI parameters, ADC values, and the T2-FLAIR mismatch sign are useful to differentiate between DMG-A and DMG-W. KEY POINTS • Diffuse midline glioma, H3K27-altered (DMG-A), showed a significantly lower normalized relative cerebral blood flow and volume compared with H3K27-wild-type counterparts (DMG-W). • T2-fluid-attenuated inversion recovery (FLAIR) mismatch sign was significantly more frequent in DMG-A compared to DMG-W. • Indicators that combined DSC parameters, ADC values, and T2-FLAIR mismatch sign, with or without age, are useful to distinguish the two tumors.
Collapse
Affiliation(s)
- Ryo Kurokawa
- Division of Neuroradiology, Department of Radiology, Michigan Medicine, 1500E Medical Center Drive, Ann Arbor, MI, 48109, USA.
| | - Mariko Kurokawa
- Division of Neuroradiology, Department of Radiology, Michigan Medicine, 1500E Medical Center Drive, Ann Arbor, MI, 48109, USA
| | - Akira Baba
- Division of Neuroradiology, Department of Radiology, Michigan Medicine, 1500E Medical Center Drive, Ann Arbor, MI, 48109, USA
| | - Yoshiaki Ota
- Division of Neuroradiology, Department of Radiology, Michigan Medicine, 1500E Medical Center Drive, Ann Arbor, MI, 48109, USA
| | - John Kim
- Division of Neuroradiology, Department of Radiology, Michigan Medicine, 1500E Medical Center Drive, Ann Arbor, MI, 48109, USA
| | - Aristides Capizzano
- Division of Neuroradiology, Department of Radiology, Michigan Medicine, 1500E Medical Center Drive, Ann Arbor, MI, 48109, USA
| | - Ashok Srinivasan
- Division of Neuroradiology, Department of Radiology, Michigan Medicine, 1500E Medical Center Drive, Ann Arbor, MI, 48109, USA
| | - Toshio Moritani
- Division of Neuroradiology, Department of Radiology, Michigan Medicine, 1500E Medical Center Drive, Ann Arbor, MI, 48109, USA
| |
Collapse
|
29
|
Kurokawa R, Umemura Y, Capizzano A, Kurokawa M, Baba A, Holmes A, Kim J, Ota Y, Srinivasan A, Moritani T. Dynamic susceptibility contrast and diffusion-weighted MRI in posterior fossa pilocytic astrocytoma and medulloblastoma. J Neuroimaging 2022; 32:511-520. [PMID: 34997668 DOI: 10.1111/jon.12962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND AND PURPOSE The utility of perfusion MRI in distinguishing between pilocytic astrocytoma (PA) and medulloblastoma (MB) is unclear. This study aimed to evaluate the diagnostic and prognostic performance of dynamic susceptibility contrast (DSC)-MRI parameters and apparent diffusion coefficient (ADC) values between PA and MB. METHODS Between January 2012 and August 2021, 49 (median, 7 years [range, 1-28 years]; 28 females) and 35 (median, 8 years [1-24 years]; 12 females) patients with pathologically confirmed PA and MB, respectively, were included. The normalized relative cerebral blood volume and flow (nrCBV and nrCBF) and mean and minimal normalized ADC (nADCmean and nADCmin) values were calculated using volume-of-interest analyses. Diagnostic performance and Pearson's correlation with progression-free survival were also evaluated. RESULTS The MB group showed a significantly higher nrCBV and nrCBF (nrCBV: 1.69 [0.93-4.23] vs. 0.95 [range, 0.37-2.28], p = .0032; nrCBF: 1.62 [0.93-3.16] vs. 1.07 [0.46-2.26], p = .0084) and significantly lower nADCmean and nADCmin (nADCmean: 0.97 [0.70-1.68] vs. 2.21 [1.44-2.80], p < .001; nADCmin: 0.50 [0.19-0.89] vs. 1.42 [0.89-2.20], p < .001) than the PA group. All parameters exhibited good diagnostic ability (accuracy >0.80) with nADCmin achieving the highest score (accuracy = 1). A moderate correlation was found between nADCmean and progression-free survival for MB (r = 0.44, p = .0084). CONCLUSIONS DSC-MRI parameters and ADC values were useful for distinguishing between PA and MB. A lower ADC indicated an unfavorable MB prognosis, but the DSC-MRI parameters did not correlate with progression-free survival in either group.
Collapse
Affiliation(s)
- Ryo Kurokawa
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Yoshie Umemura
- Department of Neurology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Aristides Capizzano
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Mariko Kurokawa
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Akira Baba
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Adam Holmes
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - John Kim
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Yoshiaki Ota
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Ashok Srinivasan
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Toshio Moritani
- Division of Neuroradiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
30
|
Liu P, Lee YZ, Aylward SR, Niethammer M. Perfusion Imaging: An Advection Diffusion Approach. IEEE TRANSACTIONS ON MEDICAL IMAGING 2021; 40:3424-3435. [PMID: 34086563 PMCID: PMC8686530 DOI: 10.1109/tmi.2021.3085828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Perfusion imaging is of great clinical importance and is used to assess a wide range of diseases including strokes and brain tumors. Commonly used approaches for the quantitative analysis of perfusion images are based on measuring the effect of a contrast agent moving through blood vessels and into tissue. Contrast-agent free approaches, for example, based on intravoxel incoherent motion and arterial spin labeling, also exist, but are so far not routinely used clinically. Existing contrast-agent-dependent methods typically rely on the estimation of the arterial input function (AIF) to approximately model tissue perfusion. These approaches neglect spatial dependencies. Further, as reliably estimating the AIF is non-trivial, different AIF estimates may lead to different perfusion measures. In this work we therefore propose PIANO, an approach that provides additional insights into the perfusion process. PIANO estimates the velocity and diffusion fields of an advection-diffusion model best explaining the contrast dynamics without using an AIF. PIANO accounts for spatial dependencies and neither requires estimating the AIF nor relies on a particular contrast agent bolus shape. Specifically, we propose a convenient parameterization of the estimation problem, a numerical estimation approach, and extensively evaluate PIANO. Simulation experiments show the robustness and effectiveness of PIANO, along with its ability to distinguish between advection and diffusion. We further apply PIANO on a public brain magnetic resonance (MR) perfusion dataset of acute stroke patients, and demonstrate that PIANO can successfully resolve velocity and diffusion field ambiguities and results in sensitive measures for the assessment of stroke, comparing favorably to conventional measures of perfusion.
Collapse
|
31
|
Dalby RB, Eskildsen SF, Videbech P, Rosenberg R, Østergaard L. Cerebral hemodynamics and capillary dysfunction in late-onset major depressive disorder. Psychiatry Res Neuroimaging 2021; 317:111383. [PMID: 34508953 DOI: 10.1016/j.pscychresns.2021.111383] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 08/22/2021] [Accepted: 08/31/2021] [Indexed: 02/06/2023]
Abstract
In major depressive disorder (MDD), perfusion changes in cortico-limbic pathways are interpreted as altered neuronal activity, but they could also signify changes in neurovascular coupling due to altered capillary function. To examine capillary function in late-onset MDD, 22 patients and 22 age- and gender-matched controls underwent perfusion MRI. We measured normalized cerebral blood flow (nCBF), cerebral blood volume (nCBV), and relative transit-time heterogeneity (RTH). Resulting brain oxygenation was estimated in terms of oxygen tension and normalized metabolic rate of oxygen (nCMRO2). Patients revealed signs of capillary dysfunction (elevated RTH) in the anterior prefrontal cortex and ventral anterior cingulate cortex bilaterally and in the left insulate cortex compared to controls, bilateral hypometabolism (parallel reductions of nCBV, nCBF, and CMRO2) but preserved capillary function in the subthalamic nucleus and globus pallidus bilaterally, and hyperactivity with preserved capillary function (increased nCBF) in the cerebellum and brainstem. Our data support that perfusion changes in deep nuclei and cerebellum reflect abnormally low and high activity, respectively, in MDD patients, but suggest that microvascular pathology affects neurovascular coupling in ventral circuits. We speculate that microvascular pathology is important for our understanding of etiology of late-onset MDD as well as infererences about resulting brain activity changes.
Collapse
Affiliation(s)
- Rikke B Dalby
- Center of Functionally Integrative Neuroscience (CFIN) / MINDLab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Centre for Psychiatric Research, Aarhus University Hospital, Risskov, Denmark; Department of Radiology, Section of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark.
| | - Simon F Eskildsen
- Center of Functionally Integrative Neuroscience (CFIN) / MINDLab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Poul Videbech
- Center for Neuropsychiatric Depression Research, Mental Health Center Glostrup, Glostrup, Denmark
| | - Raben Rosenberg
- Centre for Psychiatric Research, Aarhus University Hospital, Risskov, Denmark; Centre of Psychiatry Amager, Mental Health Services in the Capital Region of Denmark, Copenhagen, Denmark
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience (CFIN) / MINDLab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Neuroradiology Research Unit, Department of Radiology, Section of Neuroradiology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
32
|
Benzakoun J, Charron S, Turc G, Hassen WB, Legrand L, Boulouis G, Naggara O, Baron JC, Thirion B, Oppenheim C. Tissue outcome prediction in hyperacute ischemic stroke: Comparison of machine learning models. J Cereb Blood Flow Metab 2021; 41:3085-3096. [PMID: 34159824 PMCID: PMC8756479 DOI: 10.1177/0271678x211024371] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Machine Learning (ML) has been proposed for tissue fate prediction after acute ischemic stroke (AIS), with the aim to help treatment decision and patient management. We compared three different ML models to the clinical method based on diffusion-perfusion thresholding for the voxel-based prediction of final infarct, using a large MRI dataset obtained in a cohort of AIS patients prior to recanalization treatment. Baseline MRI (MRI0), including diffusion-weighted sequence (DWI) and Tmax maps from perfusion-weighted sequence, and 24-hr follow-up MRI (MRI24h) were retrospectively collected in consecutive 394 patients AIS patients (median age = 70 years; final infarct volume = 28mL). Manually segmented DWI24h lesion was considered the final infarct. Gradient Boosting, Random Forests and U-Net were trained using DWI, apparent diffusion coefficient (ADC) and Tmax maps on MRI0 as inputs to predict final infarct. Tissue outcome predictions were compared to final infarct using Dice score. Gradient Boosting had significantly better predictive performance (median [IQR] Dice Score as for median age, maybe you can replace the comma with an equal sign for consistency 0.53 [0.29-0.68]) than U-Net (0.48 [0.18-0.68]), Random Forests (0.51 [0.27-0.66]), and clinical thresholding method (0.45 [0.25-0.62]) (P < 0.001). In this benchmark of ML models for tissue outcome prediction in AIS, Gradient Boosting outperformed other ML models and clinical thresholding method and is thus promising for future decision-making.
Collapse
Affiliation(s)
- Joseph Benzakoun
- Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM U1266, Paris, France.,Department of Neuroradiology, GHU Paris Psychiatrie et Neurosciences, FHU Neurovasc, Paris, France.,Faculté de médecine, Université de Paris, Paris, France
| | - Sylvain Charron
- Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM U1266, Paris, France.,Faculté de médecine, Université de Paris, Paris, France
| | - Guillaume Turc
- Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM U1266, Paris, France.,Faculté de médecine, Université de Paris, Paris, France.,Department of Neurology, GHU Paris Psychiatrie et Neurosciences, FHU Neurovasc, Paris, France
| | - Wagih Ben Hassen
- Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM U1266, Paris, France.,Department of Neuroradiology, GHU Paris Psychiatrie et Neurosciences, FHU Neurovasc, Paris, France
| | - Laurence Legrand
- Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM U1266, Paris, France.,Department of Neuroradiology, GHU Paris Psychiatrie et Neurosciences, FHU Neurovasc, Paris, France
| | - Grégoire Boulouis
- Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM U1266, Paris, France.,Department of Neuroradiology, GHU Paris Psychiatrie et Neurosciences, FHU Neurovasc, Paris, France.,Faculté de médecine, Université de Paris, Paris, France
| | - Olivier Naggara
- Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM U1266, Paris, France.,Department of Neuroradiology, GHU Paris Psychiatrie et Neurosciences, FHU Neurovasc, Paris, France.,Faculté de médecine, Université de Paris, Paris, France
| | - Jean-Claude Baron
- Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM U1266, Paris, France.,Faculté de médecine, Université de Paris, Paris, France.,Department of Neurology, GHU Paris Psychiatrie et Neurosciences, FHU Neurovasc, Paris, France
| | | | - Catherine Oppenheim
- Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM U1266, Paris, France.,Department of Neuroradiology, GHU Paris Psychiatrie et Neurosciences, FHU Neurovasc, Paris, France.,Faculté de médecine, Université de Paris, Paris, France
| |
Collapse
|
33
|
Sudarshan VP, Reddy PK, Gubbi J, Purushothaman B. Forward Model and Deep Learning Based Iterative Deconvolution for Robust Dynamic CT Perfusion. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2021; 2021:3543-3546. [PMID: 34892004 DOI: 10.1109/embc46164.2021.9630969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Perfusion maps obtained from low-dose computed tomography (CT) data suffer from poor signal to noise ratio. To enhance the quality of the perfusion maps, several works rely on denoising the low-dose CT (LD-CT) images followed by conventional regularized deconvolution. Recent works employ deep neural networks (DNN) for learning a direct mapping between the noisy and the clean perfusion maps ignoring the convolution-based forward model. DNN-based methods are not robust to practical variations in the data that are seen in real-world applications such as stroke. In this work, we propose an iterative framework that combines the perfusion forward model with a DNN-based regularizer to obtain perfusion maps directly from the LD-CT dynamic data. To improve the robustness of the DNN, we leverage the anatomical information from the contrast-enhanced LD-CT images to learn the mapping between low-dose and standard-dose perfusion maps. Through empirical experiments, we show that our model is robust both qualitatively and quantitatively to practical perturbations in the data.
Collapse
|
34
|
Loos WS, Souza R, Andersen LB, Lebel RM, Frayne R. Extraction of a vascular function for a fully automated dynamic contrast-enhanced magnetic resonance brain image processing pipeline. Magn Reson Med 2021; 87:1561-1573. [PMID: 34708417 DOI: 10.1002/mrm.29054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 12/16/2022]
Abstract
PURPOSE To develop a deep-learning model that leverages the spatial and temporal information from dynamic contrast-enhanced magnetic resonance (DCE MR) brain imaging in order to automatically estimate a vascular function (VF) for quantitative pharmacokinetic (PK) modeling. METHODS Patients with glioblastoma multiforme were scanned post-resection approximately every 2 months using a high spatial and temporal resolution DCE MR imaging sequence ( ≈ 5 s and ≈ 2 cm3 ). A region over the transverse sinus was manually drawn in the dynamic T1-weighted images to provide a ground truth VF. The manual regions and their resulting VF curves were used to train a deep-learning model based on a 3D U-net architecture. The model concurrently utilized the spatial and temporal information in DCE MR images to predict the VF. In order to analyze the contribution of the spatial and temporal terms, different weighted combinations were examined. The manual and deep-learning predicted regions and VF curves were compared. RESULTS Forty-three patients were enrolled in this study and 155 DCE MR scans were processed. The 3D U-net was trained using a loss function that combined the spatial and temporal information with different weightings. The best VF curves were obtained when both spatial and temporal information were considered. The predicted VF curve was similar to the manual ground truth VF curves. CONCLUSION The use of spatial and temporal information improved VF curve prediction relative to when only the spatial information is used. The method generalized well for unseen data and can be used to automatically estimate a VF curve suitable for quantitative PK modeling. This method allows for a more efficient clinical pipeline and may improve automation of permeability mapping.
Collapse
Affiliation(s)
- Wallace S Loos
- Radiology and Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Seaman Family MR Research Centre, Foothills Medical Centre, Calgary, Alberta, Canada
| | - Roberto Souza
- Seaman Family MR Research Centre, Foothills Medical Centre, Calgary, Alberta, Canada.,Electrical and Software Engineering, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Linda B Andersen
- Radiology and Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Seaman Family MR Research Centre, Foothills Medical Centre, Calgary, Alberta, Canada
| | - R Marc Lebel
- Radiology and Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Seaman Family MR Research Centre, Foothills Medical Centre, Calgary, Alberta, Canada.,General Electric Healthcare, Calgary, Alberta, Canada
| | - Richard Frayne
- Radiology and Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Seaman Family MR Research Centre, Foothills Medical Centre, Calgary, Alberta, Canada
| |
Collapse
|
35
|
Ben Alaya I, Limam H, Kraiem T. Applications of artificial intelligence for DWI and PWI data processing in acute ischemic stroke: Current practices and future directions. Clin Imaging 2021; 81:79-86. [PMID: 34649081 DOI: 10.1016/j.clinimag.2021.09.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/05/2021] [Accepted: 09/22/2021] [Indexed: 11/03/2022]
Abstract
Multimodal Magnetic Resonance Imaging (MRI) techniques of Perfusion-Weighted Imaging (PWI) and Diffusion-Weighted Imaging (DWI) data are integral parts of the diagnostic workup in the acute stroke setting. The visual interpretation of PWI/DWI data is the most likely procedure to triage Acute Ischemic Stroke (AIS) patients who will access reperfusion therapy, especially in those exceeding 6 h of stroke onset. In fact, this process defines two classes of tissue: the ischemic core, which is presumed to be irreversibly damaged, visualized on DWI data and the penumbra which is the reversibly injured brain tissue around the ischemic tissue, visualized on PWI data. AIS patients with a large ischemic penumbra and limited infarction core have a high probability of benefiting from endovascular treatment. However, it is a tedious and time-consuming procedure. Consequently, it is subject to high inter- and intra-observer variability. Thus, the assessment of the potential risks and benefits of endovascular treatment is uncertain. Fast, accurate and automatic post-processing of PWI and DWI data is important for clinical diagnosis and is necessary to help the decision making for therapy. Therefore, an automated procedure that identifies stroke slices, stroke hemisphere, segments stroke regions in DWI, and measures hypoperfused tissue in PWI enhances considerably the reproducibility and the accuracy of stroke assessment. In this work, we draw an overview of several applications of Artificial Intelligence (AI) for the automation processing and their potential contributions in clinical practices. We compare the current approaches among each other's with respect to some key requirements.
Collapse
Affiliation(s)
- Ines Ben Alaya
- Tunis El Manar University, Higher Institute of Medical Technology of Tunis, Laboratory of Biophysics and Medical Technology, 1006 Tunis, Tunisia.
| | - Hela Limam
- Université de Tunis El Manar, Institut Supérieur d'Informatique, Institut Supérieur de Gestion de Tunis, Laboratoire BestMod, 1002 Tunis, Tunisie.
| | - Tarek Kraiem
- Tunis El Manar University, Higher Institute of Medical Technology of Tunis, Laboratory of Biophysics and Medical Technology, 1006 Tunis, Tunisia.
| |
Collapse
|
36
|
Anderson VC, Tagge IJ, Doud A, Li X, Springer CS, Quinn JF, Kaye JA, Wild KV, Rooney WD. DCE-MRI of Brain Fluid Barriers: In Vivo Water Cycling at the Human Choroid Plexus. Tissue Barriers 2021; 10:1963143. [PMID: 34542012 DOI: 10.1080/21688370.2021.1963143] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Metabolic deficits at brain-fluid barriers are an increasingly recognized feature of cognitive decline in older adults. At the blood-cerebrospinal fluid barrier, water is transported across the choroid plexus (CP) epithelium against large osmotic gradients via processes tightly coupled to activity of the sodium/potassium pump. Here, we quantify CP homeostatic water exchange using dynamic contrast-enhanced MRI and investigate the association of the water efflux rate constant (kco) with cognitive dysfunction in older individuals. Temporal changes in the longitudinal relaxation rate constant (R1) after contrast agent bolus injection were measured in a CP region of interest in 11 participants with mild cognitive dysfunction [CI; 73 ± 6 years] and 28 healthy controls [CN; 72 ± 7 years]. kco was determined from a modified two-site pharmacokinetic exchange analysis of the R1 time-course. Ktrans, a measure of contrast agent extravasation to the interstitial space was also determined. Cognitive function was assessed by neuropsychological test performance. kco averages 5.8 ± 2.7 s-1 in CN individuals and is reduced by 2.4 s-1 [ca. 40%] in CI subjects. Significant associations of kco with global cognition and multiple cognitive domains are observed. Ktrans averages 0.13 ± 0.07 min-1 and declines with age [-0.006 ± 0.002 min-1 yr-1], but shows no difference between CI and CN individuals or association with cognitive performance. Our findings suggest that the CP water efflux rate constant is associated with cognitive dysfunction and shows an age-related decline in later life, consistent with the metabolic disturbances that characterize brain aging.
Collapse
Affiliation(s)
- Valerie C Anderson
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Ian J Tagge
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Aaron Doud
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Xin Li
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Charles S Springer
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Joseph F Quinn
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey A Kaye
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Katherine V Wild
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - William D Rooney
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
37
|
de la Rosa E, Sima DM, Menze B, Kirschke JS, Robben D. AIFNet: Automatic vascular function estimation for perfusion analysis using deep learning. Med Image Anal 2021; 74:102211. [PMID: 34425318 DOI: 10.1016/j.media.2021.102211] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 06/25/2021] [Accepted: 08/04/2021] [Indexed: 12/30/2022]
Abstract
Perfusion imaging is crucial in acute ischemic stroke for quantifying the salvageable penumbra and irreversibly damaged core lesions. As such, it helps clinicians to decide on the optimal reperfusion treatment. In perfusion CT imaging, deconvolution methods are used to obtain clinically interpretable perfusion parameters that allow identifying brain tissue abnormalities. Deconvolution methods require the selection of two reference vascular functions as inputs to the model: the arterial input function (AIF) and the venous output function, with the AIF as the most critical model input. When manually performed, the vascular function selection is time demanding, suffers from poor reproducibility and is subject to the professionals' experience. This leads to potentially unreliable quantification of the penumbra and core lesions and, hence, might harm the treatment decision process. In this work we automatize the perfusion analysis with AIFNet, a fully automatic and end-to-end trainable deep learning approach for estimating the vascular functions. Unlike previous methods using clustering or segmentation techniques to select vascular voxels, AIFNet is directly optimized at the vascular function estimation, which allows to better recognise the time-curve profiles. Validation on the public ISLES18 stroke database shows that AIFNet almost reaches inter-rater performance for the vascular function estimation and, subsequently, for the parameter maps and core lesion quantification obtained through deconvolution. We conclude that AIFNet has potential for clinical transfer and could be incorporated in perfusion deconvolution software.
Collapse
Affiliation(s)
- Ezequiel de la Rosa
- icometrix, Leuven, Belgium; Department of Computer Science, Technical University of Munich, Munich, Germany.
| | | | - Bjoern Menze
- Department of Computer Science, Technical University of Munich, Munich, Germany
| | - Jan S Kirschke
- Neuroradiology, School of Medicine, Technical University of Munich, Munich, Germany
| | - David Robben
- icometrix, Leuven, Belgium; Medical Imaging Research Center (MIRC), KU Leuven, Leuven, Belgium; Medical Image Computing (MIC), ESAT-PSI, Department of Electrical Engineering, KU Leuven, Leuven, Belgium
| |
Collapse
|
38
|
Cerebral Macro- and Microcirculation during Ephedrine versus Phenylephrine Treatment in Anesthetized Brain Tumor Patients: A Randomized Clinical Trial Using Magnetic Resonance Imaging. Anesthesiology 2021; 135:788-803. [PMID: 34344019 DOI: 10.1097/aln.0000000000003877] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND This study compared ephedrine versus phenylephrine treatment on cerebral macro- and microcirculation, measured by cerebral blood flow, and capillary transit time heterogeneity, in anesthetized brain tumor patients. The hypothesis was that capillary transit time heterogeneity in selected brain regions is greater during phenylephrine than during ephedrine, thus reducing cerebral oxygen tension. METHODS In this single-center, double-blinded, randomized clinical trial, 24 anesthetized brain tumor patients were randomly assigned to ephedrine or phenylephrine. Magnetic resonance imaging of peritumoral and contralateral hemispheres was performed before and during vasopressor infusion. The primary endpoint was between-group difference in capillary transit time heterogeneity. Secondary endpoints included changes in cerebral blood flow, estimated oxygen extraction fraction, and brain tissue oxygen tension. RESULTS Data from 20 patients showed that mean (± SD) capillary transit time heterogeneity in the contralateral hemisphere increased during phenylephrine from 3.0 ± 0.5 to 3.2 ± 0.7 s and decreased during ephedrine from 3.1 ± 0.8 to 2.7 ± 0.7 s (difference phenylephrine versus difference ephedrine [95% CI], -0.6 [-0.9 to -0.2] s; P = 0.004). In the peritumoral region, the mean capillary transit time heterogeneity increased during phenylephrine from 4.1 ± 0.7 to 4.3 ± 0.8 s and decreased during ephedrine from 3.5 ± 0.9 to 3.3 ± 0.9 s (difference phenylephrine versus difference ephedrine [95%CI], -0.4[-0.9 to 0.1] s; P = 0.130). Cerebral blood flow (contralateral hemisphere ratio difference [95% CI], 0.3 [0.06 to 0.54]; P = 0.018; and peritumoral ratio difference [95% CI], 0.3 [0.06 to 0.54; P = 0.018) and estimated brain tissue oxygen tension (contralateral hemisphere ratio difference [95% CI], 0.34 [0.09 to 0.59]; P = 0.001; and peritumoral ratio difference [95% CI], 0.33 [0.09 to 0.57]; P = 0.010) were greater during ephedrine than phenylephrine in both regions. CONCLUSIONS Phenylephrine caused microcirculation in contralateral tissue, measured by the change in capillary transit time heterogeneity, to deteriorate compared with ephedrine, despite reaching similar mean arterial pressure endpoints. Ephedrine improved cerebral blood flow and tissue oxygenation in both brain regions and may be superior to phenylephrine in improving cerebral macro- and microscopic hemodynamics and oxygenation. EDITOR’S PERSPECTIVE
Collapse
|
39
|
Operator dependency of arterial input function in dynamic contrast-enhanced MRI. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2021; 35:105-112. [PMID: 34213687 PMCID: PMC8901481 DOI: 10.1007/s10334-021-00926-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 11/09/2022]
Abstract
Objective To investigate the effect of inter-operator variability in arterial input function (AIF) definition on kinetic parameter estimates (KPEs) from dynamic contrast-enhanced (DCE) MRI in patients with high-grade gliomas. Methods The study included 118 DCE series from 23 patients. AIFs were measured by three domain experts (DEs), and a population AIF (pop-AIF) was constructed from the measured AIFs. The DE-AIFs, pop-AIF and AUC-normalized DE-AIFs were used for pharmacokinetic analysis with the extended Tofts model. AIF-dependence of KPEs was assessed by intraclass correlation coefficient (ICC) analysis, and the impact on relative longitudinal change in Ktrans was assessed by Fleiss’ kappa (κ). Results There was a moderate to substantial agreement (ICC 0.51–0.76) between KPEs when using DE-AIFs, while AUC-normalized AIFs yielded ICC 0.77–0.95 for Ktrans, kep and ve and ICC 0.70 for vp. Inclusion of the pop-AIF did not reduce agreement. Agreement in relative longitudinal change in Ktrans was moderate (κ = 0.591) using DE-AIFs, while AUC-normalized AIFs gave substantial (κ = 0.809) agreement. Discussion AUC-normalized AIFs can reduce the variation in kinetic parameter results originating from operator input. The pop-AIF presented in this work may be applied in absence of a satisfactory measurement.
Collapse
|
40
|
Siakallis L, Sudre CH, Mulholland P, Fersht N, Rees J, Topff L, Thust S, Jager R, Cardoso MJ, Panovska-Griffiths J, Bisdas S. Longitudinal structural and perfusion MRI enhanced by machine learning outperforms standalone modalities and radiological expertise in high-grade glioma surveillance. Neuroradiology 2021; 63:2047-2056. [PMID: 34047805 PMCID: PMC8589799 DOI: 10.1007/s00234-021-02719-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/12/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE Surveillance of patients with high-grade glioma (HGG) and identification of disease progression remain a major challenge in neurooncology. This study aimed to develop a support vector machine (SVM) classifier, employing combined longitudinal structural and perfusion MRI studies, to classify between stable disease, pseudoprogression and progressive disease (3-class problem). METHODS Study participants were separated into two groups: group I (total cohort: 64 patients) with a single DSC time point and group II (19 patients) with longitudinal DSC time points (2-3). We retrospectively analysed 269 structural MRI and 92 dynamic susceptibility contrast perfusion (DSC) MRI scans. The SVM classifier was trained using all available MRI studies for each group. Classification accuracy was assessed for different feature dataset and time point combinations and compared to radiologists' classifications. RESULTS SVM classification based on combined perfusion and structural features outperformed radiologists' classification across all groups. For the identification of progressive disease, use of combined features and longitudinal DSC time points improved classification performance (lowest error rate 1.6%). Optimal performance was observed in group II (multiple time points) with SVM sensitivity/specificity/accuracy of 100/91.67/94.7% (first time point analysis) and 85.71/100/94.7% (longitudinal analysis), compared to 60/78/68% and 70/90/84.2% for the respective radiologist classifications. In group I (single time point), the SVM classifier also outperformed radiologists' classifications with sensitivity/specificity/accuracy of 86.49/75.00/81.53% (SVM) compared to 75.7/68.9/73.84% (radiologists). CONCLUSION Our results indicate that utilisation of a machine learning (SVM) classifier based on analysis of longitudinal perfusion time points and combined structural and perfusion features significantly enhances classification outcome (p value= 0.0001).
Collapse
Affiliation(s)
- Loizos Siakallis
- Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK.
| | - Carole H Sudre
- Translational Imaging Group, Centre for Medical Image Computing, University College London , London, UK.,Department of Medical Physics, University College London, London, UK
| | - Paul Mulholland
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Naomi Fersht
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Jeremy Rees
- Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, London, UK.,Department of Neurooncology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Laurens Topff
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Steffi Thust
- Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| | - Rolf Jager
- Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK.,Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, London, UK
| | - M Jorge Cardoso
- Translational Imaging Group, Centre for Medical Image Computing, University College London , London, UK
| | - Jasmina Panovska-Griffiths
- Institute for Global Health, University College London, London, UK.,The Queen's College, University of Oxford, Oxford, UK
| | - Sotirios Bisdas
- Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK.,Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, London, UK
| |
Collapse
|
41
|
Kang KM, Choi SH, Chul-Kee P, Kim TM, Park SH, Lee JH, Lee ST, Hwang I, Yoo RE, Yun TJ, Kim JH, Sohn CH. Differentiation between glioblastoma and primary CNS lymphoma: application of DCE-MRI parameters based on arterial input function obtained from DSC-MRI. Eur Radiol 2021; 31:9098-9109. [PMID: 34003350 DOI: 10.1007/s00330-021-08044-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 04/06/2021] [Accepted: 05/04/2021] [Indexed: 11/24/2022]
Abstract
OBJECTIVE This study aimed to evaluate whether arterial input functions (AIFs) obtained from dynamic susceptibility contrast (DSC)-MRI (AIFDSC) improve the reliability and diagnostic accuracy of dynamic contrast-enhanced (DCE)-derived pharmacokinetic (PK) parameters for differentiating glioblastoma from primary CNS lymphoma (PCNSL) compared with AIFs derived from DCE-MRI (AIFDCE). METHODS This retrospective study included 172 patients with glioblastoma (n = 147) and PCNSL (n = 25). All patients had undergone preoperative DSC- and DCE-MRI. The volume transfer constant (Ktrans), volume of the vascular plasma space (vp), and volume of the extravascular extracellular space (ve) were acquired using AIFDSC and AIFDCE. The relative cerebral blood volume (rCBV) was obtained from DSC-MRI. Intraclass correlation coefficients (ICC) and ROC curves were used to assess the reliability and diagnostic accuracy of individual parameters. RESULTS The mean Ktrans, vp, and ve values revealed better ICCs with AIFDSC than with AIFDCE (Ktrans, 0.911 vs 0.355; vp, 0.766 vs 0.503; ve, 0.758 vs 0.657, respectively). For differentiating all glioblastomas from PCNSL, the mean rCBV (AUC = 0.856) was more accurate than the AIFDSC-driven mean Ktrans, which had the largest AUC (0.711) among the DCE-derived parameters (p = 0.02). However, for glioblastomas with low rCBV (≤ 75th percentile of PCNSL; n = 30), the AIFDSC-driven mean Ktrans and vp were more accurate than rCBV (AUC: Ktrans, 0.807 vs rCBV, 0.515, p = 0.004; vp, 0.715 vs rCBV, p = 0.045). CONCLUSION DCE-derived PK parameters using the AIFDSC showed improved reliability and diagnostic accuracy for differentiating glioblastoma with low rCBV from PCNSL. KEY POINTS • An accurate differential diagnosis of glioblastoma and PCNSL is crucial because of different therapeutic strategies. • In contrast to the rCBV from DSC-MRI, another perfusion imaging technique, the DCE parameters for the differential diagnosis have been limited because of the low reliability of AIFs from DCE-MRI. • When we analyzed DCE-MRI data using AIFs from DSC-MRI (AIFDSC), AIFDSC-driven DCE parameters showed improved reliability and better diagnostic accuracy than rCBV for differentiating glioblastoma with low rCBV from PCNSL.
Collapse
Affiliation(s)
- Koung Mi Kang
- Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Radiology, Seoul National University Hospital, 101 Daehangno, Jongno-gu, Seoul, 110-744, Republic of Korea
| | - Seung Hong Choi
- Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea. .,Department of Radiology, Seoul National University Hospital, 101 Daehangno, Jongno-gu, Seoul, 110-744, Republic of Korea. .,Center for Nanoparticle Research, Institute for Basic Science, and School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea.
| | - Park Chul-Kee
- Department of Neurosurgery and Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Tae Min Kim
- Department of Internal Medicine and Cancer Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Joo Ho Lee
- Department of Radiation Oncology and Cancer Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Soon-Tae Lee
- Department of Neurology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Inpyeong Hwang
- Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Radiology, Seoul National University Hospital, 101 Daehangno, Jongno-gu, Seoul, 110-744, Republic of Korea
| | - Roh-Eul Yoo
- Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Radiology, Seoul National University Hospital, 101 Daehangno, Jongno-gu, Seoul, 110-744, Republic of Korea
| | - Tae Jin Yun
- Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Radiology, Seoul National University Hospital, 101 Daehangno, Jongno-gu, Seoul, 110-744, Republic of Korea
| | - Ji-Hoon Kim
- Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Radiology, Seoul National University Hospital, 101 Daehangno, Jongno-gu, Seoul, 110-744, Republic of Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Radiology, Seoul National University Hospital, 101 Daehangno, Jongno-gu, Seoul, 110-744, Republic of Korea
| |
Collapse
|
42
|
Wu D, Ren H, Li Q. Self-Supervised Dynamic CT Perfusion Image Denoising With Deep Neural Networks. IEEE TRANSACTIONS ON RADIATION AND PLASMA MEDICAL SCIENCES 2021. [DOI: 10.1109/trpms.2020.2996566] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
43
|
Eskildsen SF, Iranzo A, Stokholm MG, Stær K, Østergaard K, Serradell M, Otto M, Svendsen KB, Garrido A, Vilas D, Borghammer P, Santamaria J, Møller A, Gaig C, Brooks DJ, Tolosa E, Østergaard L, Pavese N. Impaired cerebral microcirculation in isolated REM sleep behaviour disorder. Brain 2021; 144:1498-1508. [PMID: 33880533 DOI: 10.1093/brain/awab054] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/19/2020] [Accepted: 12/09/2020] [Indexed: 01/18/2023] Open
Abstract
During the prodromal period of Parkinson's disease and other α-synucleinopathy-related parkinsonisms, neurodegeneration is thought to progressively affect deep brain nuclei, such as the locus coeruleus, caudal raphe nucleus, substantia nigra, and the forebrain nucleus basalis of Meynert. Besides their involvement in the regulation of mood, sleep, behaviour, and memory functions, these nuclei also innervate parenchymal arterioles and capillaries throughout the cortex, possibly to ensure that oxygen supplies are adjusted according to the needs of neural activity. The aim of this study was to examine whether patients with isolated REM sleep behaviour disorder, a parasomnia considered to be a prodromal phenotype of α-synucleinopathies, reveal microvascular flow disturbances consistent with disrupted central blood flow control. We applied dynamic susceptibility contrast MRI to characterize the microscopic distribution of cerebral blood flow in the cortex of 20 polysomnographic-confirmed patients with isolated REM sleep behaviour disorder (17 males, age range: 54-77 years) and 25 healthy matched controls (25 males, age range: 58-76 years). Patients and controls were cognitively tested by Montreal Cognitive Assessment and Mini Mental State Examination. Results revealed profound hypoperfusion and microvascular flow disturbances throughout the cortex in patients compared to controls. In patients, the microvascular flow disturbances were seen in cortical areas associated with language comprehension, visual processing and recognition and were associated with impaired cognitive performance. We conclude that cortical blood flow abnormalities, possibly related to impaired neurogenic control, are present in patients with isolated REM sleep behaviour disorder and associated with cognitive dysfunction. We hypothesize that pharmacological restoration of perivascular neurotransmitter levels could help maintain cognitive function in patients with this prodromal phenotype of parkinsonism.
Collapse
Affiliation(s)
- Simon F Eskildsen
- Center of Functionally Integrative Neuroscience and MINDLab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Alex Iranzo
- Department of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Morten G Stokholm
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Kristian Stær
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Karen Østergaard
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Mónica Serradell
- Department of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Marit Otto
- Department of Clinical Neurophysiology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Alicia Garrido
- Department of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Dolores Vilas
- Department of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Per Borghammer
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Joan Santamaria
- Department of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Arne Møller
- Center of Functionally Integrative Neuroscience and MINDLab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Carles Gaig
- Department of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - David J Brooks
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Aarhus, Denmark.,Translational and Clinical Research Institute, Newcastle University, England, UK
| | - Eduardo Tolosa
- Department of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain.,Parkinson disease and Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience and MINDLab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Neuroradiology Research Unit, Department of Radiology, Aarhus University Hospital, Denmark
| | - Nicola Pavese
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Aarhus, Denmark.,Translational and Clinical Research Institute, Newcastle University, England, UK
| |
Collapse
|
44
|
Brugnara G, Herweh C, Neuberger U, Bo Hansen M, Ulfert C, Mahmutoglu MA, Foltyn M, Nagel S, Schönenberger S, Heiland S, Ringleb PA, Bendszus M, Möhlenbruch M, Pfaff JAR, Vollmuth P. Dynamics of cerebral perfusion and oxygenation parameters following endovascular treatment of acute ischemic stroke. J Neurointerv Surg 2021; 14:neurintsurg-2020-017163. [PMID: 33762405 PMCID: PMC8785045 DOI: 10.1136/neurintsurg-2020-017163] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 02/06/2021] [Accepted: 02/10/2021] [Indexed: 11/03/2022]
Abstract
BACKGROUND We studied the effects of endovascular treatment (EVT) and the impact of the extent of recanalization on cerebral perfusion and oxygenation parameters in patients with acute ischemic stroke (AIS) and large vessel occlusion (LVO). METHODS Forty-seven patients with anterior LVO underwent computed tomography perfusion (CTP) before and immediately after EVT. The entire ischemic region (Tmax >6 s) was segmented before intervention, and tissue perfusion (time-to-maximum (Tmax), time-to-peak (TTP), mean transit time (MTT), cerebral blood volume (CBV), cerebral blood flow (CBF)) and oxygenation (coefficient of variation (COV), capillary transit time heterogeneity (CTH), metabolic rate of oxygen (CMRO2), oxygen extraction fraction (OEF)) parameters were quantified from the segmented area at baseline and the corresponding area immediately after intervention, as well as within the ischemic core and penumbra. The impact of the extent of recanalization (modified Treatment in Cerebral Infarction (mTICI)) on CTP parameters was assessed with the Wilcoxon test and Pearson's correlation coefficients. RESULTS The Tmax, MTT, OEF and CTH values immediately after EVT were lower in patients with complete (as compared with incomplete) recanalization, whereas CBF and COV values were higher (P<0.05) and no differences were found in other parameters. The ischemic penumbra immediately after EVT was lower in patients with complete recanalization as compared with those with incomplete recanalization (P=0.002), whereas no difference was found for the ischemic core (P=0.12). Specifically, higher mTICI scores were associated with a greater reduction of ischemic penumbra volumes (R²=-0.48 (95% CI -0.67 to -0.22), P=0.001) but not of ischemic core volumes (P=0.098). CONCLUSIONS Our study demonstrates that the ischemic penumbra is the key target of successful EVT in patients with AIS and largely determines its efficacy on a tissue level. Furthermore, we confirm the validity of the mTICI score as a surrogate parameter of interventional success on a tissue perfusion level.
Collapse
Affiliation(s)
- Gianluca Brugnara
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Baden-Württemberg, Germany
| | - Christian Herweh
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Baden-Württemberg, Germany
| | - Ulf Neuberger
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Baden-Württemberg, Germany
| | - Mikkel Bo Hansen
- Center of Functionally Integrative Neuroscience and MINDLab, Aarhus Universitet, Aarhus, Midtjylland, Denmark
| | - Christian Ulfert
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Baden-Württemberg, Germany
| | - Mustafa Ahmed Mahmutoglu
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Baden-Württemberg, Germany
| | - Martha Foltyn
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Baden-Württemberg, Germany
| | - Simon Nagel
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Baden-Württemberg, Germany
| | - Silvia Schönenberger
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Baden-Württemberg, Germany
| | - Sabine Heiland
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Baden-Württemberg, Germany
| | - Peter Arthur Ringleb
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Baden-Württemberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Baden-Württemberg, Germany
| | - Markus Möhlenbruch
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Baden-Württemberg, Germany
| | - Johannes Alex Rolf Pfaff
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Baden-Württemberg, Germany
| | - Philipp Vollmuth
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Baden-Württemberg, Germany
| |
Collapse
|
45
|
Kang C, Lee IH, Park JS, You Y, Jeong W, Ahn HJ, Min JH. Measuring global impairment of cerebral perfusion using dynamic susceptibility contrast perfusion-weighted imaging in out-of-hospital cardiac arrest survivors: A prospective preliminary study. J Neuroradiol 2020; 48:379-384. [PMID: 33340642 DOI: 10.1016/j.neurad.2020.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/25/2020] [Accepted: 12/07/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND PURPOSE This study aimed to assess the global impairment and prognostic performance of cerebral perfusions (CP) measured by dynamic susceptibility contrast perfusion-weighted imaging (DSC-PWI) in out-of-hospital cardiac arrest (OHCA) patients after sustained restoration of spontaneous circulation (ROSC). MATERIALS AND METHODS This is a single-centre, prospective observational study. OHCA patients performed DSC-PWI within 8 h after ROSC were enrolled. We quantified the CP parameters, such as cerebral blood volume (CBV), cerebral blood flow (CBF), mean transit time (MTT), time to peak (TTP), and time to maximum of the residue function (Tmax) either by normalization or arterial input function (AIF). The primary and secondary outcomes were survival to discharge and comparison of prognostic performance between CP parameters and serum neuron-specific enolase (NSE) using area under the receiver operating characteristic (AUROC) and sensitivity values. RESULTS Thirty-one patients were included in this study. CBV and TTP quantified by normalization, and MTT and Tmax quantified by AIF showed significantly higher CP values in the non-survival group (p = 0.02, 0.03, 0.02, and <0.01, respectively). Their AUROCs and 100% specific sensitivities were 0.74/25.0%, 0.60/33.3%, 0.75/56.3%, and 0.79/43.8%, respectively. MTT quantified by AIF showed sensitivity in predicting mortality at an early stage of PCA care, comparable with NSE. CONCLUSION Hyperaemia and delayed CP were generally observed in OHCA patients regardless of outcomes. MTT and Tmax quantified by AIF have prognostic performance in predicting mortality, comparable with NSE. Further prospective multicentre studies are required to confirm our results.
Collapse
Affiliation(s)
- Changshin Kang
- Department of Emergency Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea; Department of Emergency Medicine, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - In Ho Lee
- Department of Radiology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jung Soo Park
- Department of Emergency Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea; Department of Emergency Medicine, College of Medicine, Chungnam National University, Daejeon, Republic of Korea.
| | - Yeonho You
- Department of Emergency Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Wonjoon Jeong
- Department of Emergency Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Hong Joon Ahn
- Department of Emergency Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea; Department of Emergency Medicine, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jin Hong Min
- Department of Emergency Medicine, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
46
|
Tönnes C, Janssen S, Golla AK, Uhrig T, Chung K, Schad LR, Zöllner FG. Deterministic Arterial Input Function selection in DCE-MRI for automation of quantitative perfusion calculation of colorectal cancer. Magn Reson Imaging 2020; 75:116-123. [PMID: 32987123 DOI: 10.1016/j.mri.2020.09.009] [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: 04/06/2020] [Revised: 08/27/2020] [Accepted: 09/08/2020] [Indexed: 10/23/2022]
Abstract
Development of a deterministic algorithm for automated detection of the Arterial Input Function (AIF) in DCE-MRI of colorectal cancer. Using a filter pipeline to determine the AIF region of interest. Comparison to algorithms from literature with mean squared error and quantitative perfusion parameter Ktrans. The AIF found by our algorithm has a lower mean squared error (0.0022 ± 0.0021) in reference to the manual annotation than comparable algorithms. The error of Ktrans (21.52 ± 17.2%) is lower than that of other algorithms. Our algorithm generates reproducible results and thus supports a robust and comparable perfusion analysis.
Collapse
Affiliation(s)
- Christian Tönnes
- Computer Assisted Clinical Medicine, Mannheim Institute for Intelligent Systems in Medicine, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany.
| | - Sonja Janssen
- Department for Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| | - Alena-Kathrin Golla
- Computer Assisted Clinical Medicine, Mannheim Institute for Intelligent Systems in Medicine, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| | - Tanja Uhrig
- Computer Assisted Clinical Medicine, Mannheim Institute for Intelligent Systems in Medicine, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| | - Khanlian Chung
- Computer Assisted Clinical Medicine, Mannheim Institute for Intelligent Systems in Medicine, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| | - Lothar R Schad
- Computer Assisted Clinical Medicine, Mannheim Institute for Intelligent Systems in Medicine, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| | - Frank Gerrit Zöllner
- Computer Assisted Clinical Medicine, Mannheim Institute for Intelligent Systems in Medicine, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| |
Collapse
|
47
|
Novel Estimation of Penumbra Zone Based on Infarct Growth Using Machine Learning Techniques in Acute Ischemic Stroke. J Clin Med 2020; 9:jcm9061977. [PMID: 32599812 PMCID: PMC7355454 DOI: 10.3390/jcm9061977] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/15/2020] [Accepted: 06/22/2020] [Indexed: 12/20/2022] Open
Abstract
While the penumbra zone is traditionally assessed based on perfusion-diffusion mismatch, it can be assessed based on machine learning (ML) prediction of infarct growth. The purpose of this work was to develop and validate an ML method for the prediction of infarct growth distribution and volume, in cases of successful (SR) and unsuccessful recanalization (UR). Pre-treatment perfusion-weighted, diffusion-weighted imaging (DWI) data, and final infarct lesions annotated from day-7 DWI from patients with middle cerebral artery occlusion were utilized to develop and validate two ML models for prediction of tissue fate. SR and UR models were developed from data in patients with modified treatment in cerebral infarction (mTICI) scores of 2b-3 and 0-2a, respectively. When compared to manual infarct annotation, ML-based infarct volume predictions resulted in an intraclass correlation coefficient (ICC) of 0.73 (95% CI = 0.31-0.91, p < 0.01) for UR, and an ICC of 0.87 (95% CI = 0.73-0.94, p < 0.001) for SR. Favorable outcomes for mismatch presence and absence in SR were 50% and 36%, respectively, while they were 61%, 56%, and 25%, respectively, for the low, intermediate, and high infarct growth groups. The presented method can offer novel and alternative insights into selecting patients for recanalization therapy and predicting functional outcome.
Collapse
|
48
|
Schmidt MA, Engelhorn T, Lang S, Luecking H, Hoelter P, Froehlich K, Ritt P, Maler JM, Kuwert T, Kornhuber J, Doerfler A. DSC Brain Perfusion Using Advanced Deconvolution Models in the Diagnostic Work-up of Dementia and Mild Cognitive Impairment: A Semiquantitative Comparison with HMPAO-SPECT-Brain Perfusion. J Clin Med 2020; 9:jcm9061800. [PMID: 32527014 PMCID: PMC7356248 DOI: 10.3390/jcm9061800] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/08/2020] [Accepted: 06/08/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND SPECT (single-photon emission-computed tomography) is used for the detection of hypoperfusion in cognitive impairment and dementia but is not widely available and related to radiation dose exposure. We compared the performance of DSC (dynamic susceptibility contrast) perfusion using semi- and fully adaptive deconvolution models to HMPAO-SPECT (99mTc-hexamethylpropyleneamine oxime-SPECT). MATERIAL AND METHODS Twenty-seven patients with dementia of different subtypes including frontotemporal dementia (FTD) and mild cognitive impairment (MCI) received a multimodal diagnostic work-up including DSC perfusion at a clinical 3T high-field scanner and HMPAO-SPECT. Nineteen healthy control individuals received DSC perfusion. For calculation of the hemodynamic parameter maps, oscillation-index standard truncated singular value decomposition (oSVD, semi-adaptive) as well as Bayesian parameter estimation (BAY, fully adaptive) were performed. RESULTS Patients showed decreased cortical perfusion in the left frontal lobe compared to controls (relative cerebral blood volume corrected, rBVc: 0.37 vs 0.27, p = 0.048, adjusted for age and sex). Performance of rBVc (corrected for T1 effects) was highest compared to SPECT for detection of frontal hypoperfusion (sensitivity 83%, specificity 80% for oSVD and BAY, area under curve (AUC) = 0.833 respectively, p < 0.05) in FTD and MCI. For nonleakage-corrected rBV and for rBF (relative cerebral blood flow), sensitivity of frontal hypoperfusion was above 80% for oSVD and for BAY (rBV: sensitivity 83%, specificity 75%, AUC = 0.908 for oSVD and 0.917 for BAY, p < 0.05 respectively; rBF: sensitivity 83%, specificity 65%, AUC = 0.825, p < 0.05 for oSVD). CONCLUSION Advanced deconvolution DSC can reliably detect pathological perfusion alterations in FTD and MCI. Hence, this widely accessible technique has the potential to improve the diagnosis of dementia and MCI as part of an interdisciplinary multimodal imaging work-up. Advances in knowledge: Advanced DSC perfusion has a high potential in the work-up of suspected dementia and correlates with SPECT brain perfusion results in dementia and MCI.
Collapse
Affiliation(s)
- Manuel A. Schmidt
- Departments of Neuroradiology, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054 Erlangen, Germany; (T.E.); (S.L.); (H.L.); (P.H.); (A.D.)
- Correspondence: ; Tel.: +49-9131-85-44821
| | - Tobias Engelhorn
- Departments of Neuroradiology, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054 Erlangen, Germany; (T.E.); (S.L.); (H.L.); (P.H.); (A.D.)
| | - Stefan Lang
- Departments of Neuroradiology, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054 Erlangen, Germany; (T.E.); (S.L.); (H.L.); (P.H.); (A.D.)
| | - Hannes Luecking
- Departments of Neuroradiology, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054 Erlangen, Germany; (T.E.); (S.L.); (H.L.); (P.H.); (A.D.)
| | - Philip Hoelter
- Departments of Neuroradiology, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054 Erlangen, Germany; (T.E.); (S.L.); (H.L.); (P.H.); (A.D.)
| | - Kilian Froehlich
- Departments of Neurology, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054 Erlangen, Germany;
| | - Philipp Ritt
- Department of Nuclear Medicine, Friedrich-Alexander-University Erlangen-Nuremberg, Ulmenweg 18, 91054 Erlangen, Germany; (P.R.); (T.K.)
| | - Juan Manuel Maler
- Departments of Psychiatry, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054 Erlangen, Germany; (J.M.M.); (J.K.)
| | - Torsten Kuwert
- Department of Nuclear Medicine, Friedrich-Alexander-University Erlangen-Nuremberg, Ulmenweg 18, 91054 Erlangen, Germany; (P.R.); (T.K.)
| | - Johannes Kornhuber
- Departments of Psychiatry, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054 Erlangen, Germany; (J.M.M.); (J.K.)
| | - Arnd Doerfler
- Departments of Neuroradiology, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054 Erlangen, Germany; (T.E.); (S.L.); (H.L.); (P.H.); (A.D.)
| |
Collapse
|
49
|
Nielsen RB, Parbo P, Ismail R, Dalby R, Tietze A, Brændgaard H, Gottrup H, Brooks DJ, Østergaard L, Eskildsen SF. Impaired perfusion and capillary dysfunction in prodromal Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2020; 12:e12032. [PMID: 32490139 PMCID: PMC7241262 DOI: 10.1002/dad2.12032] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/21/2020] [Accepted: 02/24/2020] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Cardiovascular disease increases the risk of developing Alzheimer's disease (AD), and growing evidence suggests an involvement of cerebrovascular pathology in AD. Capillary dysfunction, a condition in which capillary flow disturbances rather than arterial blood supply limit brain oxygen extraction, could represent an overlooked vascular contributor to neurodegeneration. We examined whether cortical capillary transit-time heterogeneity (CTH), an index of capillary dysfunction, is elevated in amyloid-positive patients with mild cognitive impairment (prodromal AD [pAD]). METHODS We performed structural and perfusion weighted MRI in 22 pAD patients and 21 healthy controls. RESULTS We found hypoperfusion, reduced blood volume, and elevated CTH in the parietal and frontal cortices of pAD-patients compared to controls, while only the precuneus showed focal cortical atrophy. DISCUSSION We propose that microvascular flow disturbances antedate cortical atrophy and may limit local tissue oxygenation in pAD. We speculate that capillary dysfunction contributes to the development of neurodegeneration in AD.
Collapse
Affiliation(s)
- Rune B. Nielsen
- Center of Functionally Integrative NeuroscienceAarhus UniversityAarhusDenmark
| | - Peter Parbo
- Department of Nuclear Medicine and PET CentreAarhus University HospitalAarhusDenmark
| | - Rola Ismail
- Department of Nuclear Medicine and PET CentreAarhus University HospitalAarhusDenmark
| | - Rikke Dalby
- Center of Functionally Integrative NeuroscienceAarhus UniversityAarhusDenmark
- Department of NeuroradiologyAarhus University HospitalAarhusDenmark
| | - Anna Tietze
- Charité, UniversitätsmedizinInstitute of NeuroradiologyBerlinGermany
| | - Hans Brændgaard
- Dementia ClinicDepartment of NeurologyAarhus University HospitalAarhusDenmark
| | - Hanne Gottrup
- Dementia ClinicDepartment of NeurologyAarhus University HospitalAarhusDenmark
| | - David J. Brooks
- Department of Nuclear Medicine and PET CentreAarhus University HospitalAarhusDenmark
- Division of NeuroscienceDepartment of MedicineImperial College LondonLondonUK
- Division of NeuroscienceNewcastle UniversityNewcastle upon TyneUK
| | - Leif Østergaard
- Center of Functionally Integrative NeuroscienceAarhus UniversityAarhusDenmark
- Department of Nuclear Medicine and PET CentreAarhus University HospitalAarhusDenmark
| | - Simon F. Eskildsen
- Center of Functionally Integrative NeuroscienceAarhus UniversityAarhusDenmark
| |
Collapse
|
50
|
Perfusion and diffusion in meningioma tumors: a preliminary multiparametric analysis with Dynamic Susceptibility Contrast and IntraVoxel Incoherent Motion MRI. Magn Reson Imaging 2020; 67:69-78. [DOI: 10.1016/j.mri.2019.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 11/15/2019] [Accepted: 12/05/2019] [Indexed: 12/19/2022]
|