Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Magn Reson Med ; 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39119838

ABSTRACT

PURPOSE: To evaluate reproducibility and interlobar agreement of intravoxel incoherent motion (IVIM) quantification in the liver across field strengths and MR scanners with different gradient hardware. METHODS: Cramer-Rao lower bound optimization was performed to determine optimized monopolar and motion-robust 2D (b-value and first-order motion moment [M1]) IVIM-DWI acquisitions. Eleven healthy volunteers underwent diffusion MRI of the liver, where each optimized acquisition was obtained five times across three MRI scanners. For each data set, IVIM estimates (diffusion coefficient (D), pseudo-diffusion coefficients ( d 1 * $$ {d}_1^{\ast } $$ and d 2 * $$ {d}_2^{\ast } $$ ), blood velocity SDs (Vb1 and Vb2), and perfusion fractions [f1 and f2]) were obtained in the right and left liver lobes using two signal models (pseudo-diffusion and M1-dependent physical) with and without T2 correction (fc1 and fc2) and three fitting techniques (tri-exponential region of interest-based full and segmented fitting and blood velocity SD distribution fitting). Reproducibility and interlobar agreement were compared across methods using within-subject and pairwise coefficients of variation (CVw and CVp), paired sample t-tests, and Bland-Altman analysis. RESULTS: Using a combination of motion-robust 2D (b-M1) data acquisition, M1-dependent physical signal modeling with T2 correction, and blood velocity SD distribution fitting, multiscanner reproducibility with median CVw = 5.09%, 11.3%, 9.20%, 14.2%, and 12.6% for D, Vb1, Vb2, fc1, and fc2, respectively, and interlobar agreement with CVp = 8.14%, 11.9%, 8.50%, 49.9%, and 42.0%, respectively, was achieved. CONCLUSION: Recently proposed advanced IVIM acquisition, signal modeling, and fitting techniques may facilitate reproducible IVIM quantification in the liver, as needed for establishment of IVIM-based quantitative biomarkers for detection, staging, and treatment monitoring of diseases.

2.
Magn Reson Med ; 2024 Jun 23.
Article in English | MEDLINE | ID: mdl-38923009

ABSTRACT

PURPOSE: Quantitative T1 mapping has the potential to replace biopsy for noninvasive diagnosis and quantitative staging of chronic liver disease. Conventional T1 mapping methods are confounded by fat and B 1 + $$ {B}_1^{+} $$ inhomogeneities, resulting in unreliable T1 estimations. Furthermore, these methods trade off spatial resolution and volumetric coverage for shorter acquisitions with only a few images obtained within a breath-hold. This work proposes a novel, volumetric (3D), free-breathing T1 mapping method to account for multiple confounding factors in a single acquisition. THEORY AND METHODS: Free-breathing, confounder-corrected T1 mapping was achieved through the combination of non-Cartesian imaging, magnetization preparation, chemical shift encoding, and a variable flip angle acquisition. A subspace-constrained, locally low-rank image reconstruction algorithm was employed for image reconstruction. The accuracy of the proposed method was evaluated through numerical simulations and phantom experiments with a T1/proton density fat fraction phantom at 3.0 T. Further, the feasibility of the proposed method was investigated through contrast-enhanced imaging in healthy volunteers, also at 3.0 T. RESULTS: The method showed excellent agreement with reference measurements in phantoms across a wide range of T1 values (200 to 1000 ms, slope = 0.998 (95% confidence interval (CI) [0.963 to 1.035]), intercept = 27.1 ms (95% CI [0.4 54.6]), r2 = 0.996), and a high level of repeatability. In vivo imaging studies demonstrated moderate agreement (slope = 1.099 (95% CI [1.067 to 1.132]), intercept = -96.3 ms (95% CI [-82.1 to -110.5]), r2 = 0.981) compared to saturation recovery-based T1 maps. CONCLUSION: The proposed method produces whole-liver, confounder-corrected T1 maps through simultaneous estimation of T1, proton density fat fraction, and B 1 + $$ {B}_1^{+} $$ in a single, free-breathing acquisition and has excellent agreement with reference measurements in phantoms.

3.
MAGMA ; 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38896407

ABSTRACT

In this paper, we review the value of phantoms for body MRI in the context of their uses for quantitative MRI methods research, clinical trials, and clinical imaging. Certain uses of phantoms are common throughout the body MRI community, including measuring bias, assessing reproducibility, and training. In addition to these uses, phantoms in body MRI methods research are used for novel methods development and the design of motion compensation and mitigation techniques. For clinical trials, phantoms are an essential part of quality management strategies, facilitating the conduct of ethically sound, reliable, and regulatorily compliant clinical research of both novel MRI methods and therapeutic agents. In the clinic, phantoms are used for development of protocols, mitigation of cost, quality control, and radiotherapy. We briefly review phantoms developed for quantitative body MRI, and finally, we review open questions regarding the most effective use of a phantom for body MRI.

4.
Magn Reson Med ; 92(4): 1743-1754, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38725136

ABSTRACT

PURPOSE: To investigate the effect of particle size on liver R 2 * $$ {\mathrm{R}}_2^{\ast } $$ by Monte Carlo simulation and phantom studies at both 1.5 T and 3.0 T. METHODS: Two kinds of particles (i.e., iron sphere and fat droplet) with varying sizes were considered separately in simulation and phantom studies. MRI signals were synthesized and analyzed for predicting R 2 * $$ {\mathrm{R}}_2^{\ast } $$ , based on simulations by incorporating virtual liver model, particle distribution, magnetic field generation, and proton movement into phase accrual. In the phantom study, iron-water and fat-water phantoms were constructed, and each phantom contained 15 separate vials with combinations of five particle concentrations and three particle sizes. R 2 * $$ {\mathrm{R}}_2^{\ast } $$ measurements in the phantom were made at both 1.5 T and 3.0 T. Finally, differences in R 2 * $$ {\mathrm{R}}_2^{\ast } $$ predictions or measurements were evaluated across varying particle sizes. RESULTS: In the simulation study, strong linear and positively correlated relationships were observed between R 2 * $$ {\mathrm{R}}_2^{\ast } $$ predictions and particle concentrations across varying particle sizes and magnetic field strengths ( r ≥ 0.988 $$ r\ge 0.988 $$ ). The relationships were affected by iron sphere size ( p < 0.001 $$ p<0.001 $$ ), where smaller iron sphere size yielded higher predicted R 2 * $$ {\mathrm{R}}_2^{\ast } $$ , whereas fat droplet size had no effect on R 2 * $$ {\mathrm{R}}_2^{\ast } $$ predictions ( p ≥ 0.617 $$ p\ge 0.617 $$ ) for constant total fat concentration. Similarly, the phantom study showed that R 2 * $$ {\mathrm{R}}_2^{\ast } $$ measurements were relatively sensitive to iron sphere size ( p ≤ 0.004 $$ p\le 0.004 $$ ) unlike fat droplet size ( p ≥ 0.223 $$ p\ge 0.223 $$ ). CONCLUSION: Liver R 2 * $$ {\mathrm{R}}_2^{\ast } $$ is affected by iron sphere size, but is relatively unaffected by fat droplet size. These findings may lead to an improved understanding of the underlying mechanisms of R 2 * $$ {\mathrm{R}}_2^{\ast } $$ relaxometry in vivo, and enable improved quantitative MRI phantom design.


Subject(s)
Computer Simulation , Liver , Magnetic Resonance Imaging , Monte Carlo Method , Particle Size , Phantoms, Imaging , Magnetic Resonance Imaging/methods , Liver/diagnostic imaging , Humans
5.
J Magn Reson Imaging ; 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38662618

ABSTRACT

BACKGROUND: Recent multicenter, multivendor MRI-based R2* vs. liver iron concentration (LIC) calibrations (i.e., MCMV calibrations) may facilitate broad clinical dissemination of R2*-based LIC quantification. However, these calibrations are based on a centralized offline R2* reconstruction, and their applicability with vendor-provided R2* maps is unclear. PURPOSE: To determine R2* ranges of agreement between the centralized and three MRI vendors' R2* reconstructions. STUDY TYPE: Prospective. SUBJECTS: Two hundred and seven subjects (mean age 37.6 ± 19.6 years; 117 male) with known or suspected iron overload from four academic medical centers. FIELD STRENGTH/SEQUENCE: Standardized multiecho spoiled gradient echo sequence at 1.5 T and 3.0 T for R2* mapping and a multiple spin-echo sequence at 1.5 T for LIC quantification. MRI vendors: GE Healthcare, Philips Healthcare, and Siemens Healthineers. ASSESSMENT: R2* maps were generated using both the centralized and vendor reconstructions, and ranges of agreement were determined. R2*-LIC linear calibrations were determined for each site, field strength, and reconstruction and compared with the MCMV calibrations. STATISTICAL TESTS: Bland-Altman analysis to determine ranges of agreement. Linear regression, analysis of covariance F tests, and Tukey's multiple comparison testing to assess reproducibility of calibrations across sites and vendors. A P value <0.05 was considered significant. RESULTS: The upper limits of R2* ranges of agreement were approximately 500, 375, and 330 s-1 for GE, Philips, and Siemens reconstructions, respectively, at 1.5 T and approximately 700 and 800 s-1 for GE and Philips, respectively, at 3.0 T. Within the R2* ranges of agreement, vendor R2*-LIC calibrations demonstrated high reproducibility (no significant differences between slopes or intercepts; P ≥ 0.06) and agreed with the MCMV calibrations (overlapping 95% confidence intervals). DATA CONCLUSION: Based on the determined upper limits, R2* measurements obtained from vendor-provided R2* maps may be reliably and practically used to quantify LIC less than approximately 8-13 mg/g using the MCMV calibrations and similar acquisition parameters as this study. EVIDENCE LEVEL: 1 TECHNICAL EFFICACY: Stage 3.

6.
Magn Reson Imaging ; 110: 161-169, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38641212

ABSTRACT

BACKGROUND: Diffusion weighted imaging (DWI) with optimized motion-compensated gradient waveforms reduces signal dropouts in the liver and pancreas caused by cardiovascular-associated motion, however its precision is unknown. We hypothesized that DWI with motion-compensated DW gradient waveforms would improve apparent diffusion coefficient (ADC)-repeatability and inter-reader reproducibility compared to conventional DWI in these organs. METHODS: In this IRB-approved, prospective, single center study, subjects recruited between October 2019 and March 2020 were scanned twice on a 3 T scanner, with repositioning between test and retest. Each scan included two respiratory-triggered DWI series with comparable acquisition time: 1) conventional (monopolar) 2) motion- compensated diffusion gradients. Three readers measured ADC values. One-way ANOVA, Bland-Altman analysis were used for statistical analysis. RESULTS: Eight healthy participants (4 male/4 female), with a mean age of 29 ± 4 years, underwent the liver and pancreas MRI protocol. Four patients with liver metastases (2 male/2 female) with a mean age of 58 ± 5 years underwent the liver MRI protocol. In healthy participants, motion-compensated DWI outperformed conventional DWI with mean repeatability coefficient of 0.14 × 10-3 (CI:0.12-0.17) vs. 0.31 × 10-3 (CI:0.27-0.37) mm2/s for liver, and 0.11 × 10-3 (CI:0.08-0.15) vs. 0.34 × 10-3 (CI:0.27-0.49) mm2/s for pancreas; and with mean reproducibility coefficient of 0.20 × 10-3 (CI:0.18-0.23) vs. 0.51 × 10-3 (CI:0.46-0.58) mm2/s for liver, and 0.16 × 10-3 (CI:0.13-0.20) vs. 0.42 × 10-3 (CI:0.34-0.52) mm2/s for pancreas. In patients, improved repeatability was observed for motion-compensated DWI in comparison to conventional with repeatability coefficient of 0.51 × 10- 3 mm2/s (CI:0.35-0.89) vs. 0.70 × 10-3 mm2/s (CI:0.49-1.20). CONCLUSION: Motion-compensated DWI enhances the precision of ADC measurements in the liver and pancreas compared to conventional DWI.


Subject(s)
Diffusion Magnetic Resonance Imaging , Liver , Motion , Pancreas , Humans , Male , Female , Diffusion Magnetic Resonance Imaging/methods , Pancreas/diagnostic imaging , Adult , Liver/diagnostic imaging , Reproducibility of Results , Prospective Studies , Middle Aged , Image Processing, Computer-Assisted/methods , Liver Neoplasms/diagnostic imaging , Image Interpretation, Computer-Assisted/methods
7.
MAGMA ; 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38349454

ABSTRACT

OBJECTIVE: Performance assessments of quantitative determinations of proton density fat fraction (PDFF) have largely focused on the range between 0 and 50%. We evaluate PDFF in a two-site phantom study across the full 0-100% PDFF range. MATERIALS AND METHODS: We used commercially available 3D chemical-shift-encoded water-fat MRI sequences from three MRI system vendors at 1.5T and 3T and conducted the study across two sites. A spherical phantom housing 18 vials spanning the full 0-100% PDFF range was used. Data at each site were acquired using default parameters to determine same-day and different-day intra-scanner repeatability, and inter-system and inter-site reproducibility, in addition to linear regression between reference and measured PDFF values. RESULTS: Across all systems, results demonstrated strong linearity and minimal bias. For 1.5T systems, a pooled slope of 0.99 with a 95% confidence interval (CI) of 0.981-0.997 and a pooled intercept of 0.61% PDFF with a 95% CI of 0.17-1.04 were obtained. Results for pooled 3T data included a slope of 1.00 (95% CI 0.995-1.005) and an intercept of 0.69% PDFF (95% CI 0.39-0.97). Inter-site and inter-system reproducibility coefficients ranged from 2.9 to 6.2 (in units of PDFF), while intra-scanner same-day and different-day repeatability ranged from 0.6 to 7.8. DISCUSSION: PDFF across the 0-100% range can be reliably estimated using current commercial offerings at 1.5T and 3T.

8.
Magn Reson Med ; 91(5): 2172-2187, 2024 May.
Article in English | MEDLINE | ID: mdl-38174431

ABSTRACT

PURPOSE: The objective was to develop a fully automated algorithm that generates confidence maps to identify regions valid for analysis of quantitative proton density fat fraction (PDFF) and R 2 * $$ {R}_2^{\ast } $$ maps of the liver, generated with chemical shift-encoded MRI (CSE-MRI). Confidence maps are urgently needed for automated quality assurance, particularly with the emergence of automated segmentation and analysis algorithms. METHODS: Confidence maps for both PDFF and R 2 * $$ {R}_2^{\ast } $$ maps are generated based on goodness of fit, measured by normalized RMS error between measured complex signals and the CSE-MRI signal model. Based on Cramér-Rao lower bound and Monte-Carlo simulations, normalized RMS error threshold criteria were developed to identify unreliable regions in quantitative maps. Simulation, phantom, and in vivo clinical studies were included. To analyze the clinical data, a board-certified radiologist delineated regions of interest (ROIs) in each of the nine liver segments for PDFF and R 2 * $$ {R}_2^{\ast } $$ analysis in consecutive clinical CSE-MRI data sets. The percent area of ROIs in areas deemed unreliable by confidence maps was calculated to assess the impact of confidence maps on real-world clinical PDFF and R 2 * $$ {R}_2^{\ast } $$ measurements. RESULTS: Simulations and phantom studies demonstrated that the proposed algorithm successfully excluded regions with unreliable PDFF and R 2 * $$ {R}_2^{\ast } $$ measurements. ROI analysis by the radiologist revealed that 2.6% and 15% of the ROIs were placed in unreliable areas of PDFF and R 2 * $$ {R}_2^{\ast } $$ maps, as identified by confidence maps. CONCLUSION: A proposed confidence map algorithm that identifies reliable areas of PDFF and R 2 * $$ {R}_2^{\ast } $$ measurements from CSE-MRI acquisitions was successfully developed. It demonstrated technical and clinical feasibility.


Subject(s)
Liver , Protons , Reproducibility of Results , Liver/diagnostic imaging , Phantoms, Imaging , Magnetic Resonance Imaging
SELECTION OF CITATIONS
SEARCH DETAIL