Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Magn Reson Med ; 92(6): 2404-2419, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39030953

RESUMEN

PURPOSE: To develop a SNR enhancement method for CEST imaging using a denoising convolutional autoencoder (DCAE) and compare its performance with state-of-the-art denoising methods. METHOD: The DCAE-CEST model encompasses an encoder and a decoder network. The encoder learns features from the input CEST Z-spectrum via a series of one-dimensional convolutions, nonlinearity applications, and pooling. Subsequently, the decoder reconstructs an output denoised Z-spectrum using a series of up-sampling and convolution layers. The DCAE-CEST model underwent multistage training in an environment constrained by Kullback-Leibler divergence, while ensuring data adaptability through context learning using Principal Component Analysis-processed Z-spectrum as a reference. The model was trained using simulated Z-spectra, and its performance was evaluated using both simulated data and in vivo data from an animal tumor model. Maps of amide proton transfer (APT) and nuclear Overhauser enhancement (NOE) effects were quantified using the multiple-pool Lorentzian fit, along with an apparent exchange-dependent relaxation metric. RESULTS: In digital phantom experiments, the DCAE-CEST method exhibited superior performance, surpassing existing denoising techniques, as indicated by the peak SNR and Structural Similarity Index. Additionally, in vivo data further confirm the effectiveness of the DCAE-CEST in denoising the APT and NOE maps when compared with other methods. Although no significant difference was observed in APT between tumors and normal tissues, there was a significant difference in NOE, consistent with previous findings. CONCLUSION: The DCAE-CEST can learn the most important features of the CEST Z-spectrum and provide the most effective denoising solution compared with other methods.


Asunto(s)
Aprendizaje Profundo , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Fantasmas de Imagen , Relación Señal-Ruido , Animales , Imagen por Resonancia Magnética/métodos , Procesamiento de Imagen Asistido por Computador/métodos , Ratones , Algoritmos , Análisis de Componente Principal
2.
bioRxiv ; 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38895366

RESUMEN

Purpose: To develop a SNR enhancement method for chemical exchange saturation transfer (CEST) imaging using a denoising convolutional autoencoder (DCAE), and compare its performance with state-of-the-art denoising methods. Method: The DCAE-CEST model encompasses an encoder and a decoder network. The encoder learns features from the input CEST Z-spectrum via a series of 1D convolutions, nonlinearity applications and pooling. Subsequently, the decoder reconstructs an output denoised Z-spectrum using a series of up-sampling and convolution layers. The DCAE-CEST model underwent multistage training in an environment constrained by Kullback-Leibler divergence, while ensuring data adaptability through context learning using Principal Component Analysis processed Z-spectrum as a reference. The model was trained using simulated Z-spectra, and its performance was evaluated using both simulated data and in-vivo data from an animal tumor model. Maps of amide proton transfer (APT) and nuclear Overhauser enhancement (NOE) effects were quantified using the multiple-pool Lorentzian fit, along with an apparent exchange-dependent relaxation metric. Results: In digital phantom experiments, the DCAE-CEST method exhibited superior performance, surpassing existing denoising techniques, as indicated by the peak SNR and Structural Similarity Index. Additionally, in vivo data further confirms the effectiveness of the DCAE-CEST in denoising the APT and NOE maps when compared to other methods. While no significant difference was observed in APT between tumors and normal tissues, there was a significant difference in NOE, consistent with previous findings. Conclusion: The DCAE-CEST can learn the most important features of the CEST Z-spectrum and provide the most effective denoising solution compared to other methods.

3.
Quant Imaging Med Surg ; 14(3): 2603-2613, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38545036

RESUMEN

Background: Neurodegeneration has been suggested to be associated with cerebral small vessel disease (CSVD). The association between different CSVD imaging markers and the extent of neurodegeneration could be indirectly confirmed by examining the relationship between CSVD imaging markers and the hippocampal amide proton transfer (APT) values. The associations between hippocampal APT values with CSVD imaging markers and CSVD total load need to be further validated. The aim of this study was to investigate potential variations in hippocampal APT values among individuals with CSVD imaging markers and varying degrees of CSVD total burden. Methods: A cross-sectional study (retrospective analysis of prospectively-acquired data) was conducted at Nanxishan Hospital of Guangxi Zhuang Autonomous Region. From May 2020 to June 2021, 165 individuals (age, 40-76 years; male/female, 103/62) were included in this study. The inclusion criteria for the participants were as follows: The presence of lacunar infarction (LI), and/or cerebral microbleed (CMB); moderate-to-severe enlarged perivascular space (EPVS) (>20); deep white matter hyperintensity (WMH) > Fazekas 2 or periventricular WMH > Fazekas. The exclusion criteria comprised the following: History of craniocerebral operation; Cases with significant pathology incidentally identified during magnetic resonance (MR) scan; Drug or alcohol abuse. The differences of hippocampal APT values between CSVD imaging makers presence or absence groups and different CSVD total burden groups were compared using independent t-test and one-way analysis of variance (ANOVA). The correlations between APT values and CSVD imaging markers were analyzed using Pearson correlation analysis. A mediation analysis model was used to investigate the mediating effect of the hippocampal APT values in the association between CSVD total loads and Montreal Cognitive Assessment (MoCA) score was assessed. Results: The hippocampal APT values among different CSVD total load groups were significantly different (P<0.001). The hippocampal APT values were significantly different between the imaging markers presence and absence groups. The P values for the LI, WMH EPVS, and CMB presence or absence groups were <0.001, <0.001, 0.034, and 0.002, respectively. The hippocampal APT values were significantly correlated with CMB (P<0.01), LI (P<0.01) and WMH (P<0.01). The mediation models demonstrated that the APT values of the hippocampus partially mediated the association between CSVD total load and MoCA score, the proportion of mediation attributable was calculated as 17.50%. Conclusions: Hippocampal APT values were associated with CSVD imaging markers and total burden. Hippocampal APT values may serve as a biomarker for the early detection of neurodegeneration in CSVD patients.

4.
Radiol Phys Technol ; 17(1): 93-102, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37897684

RESUMEN

The aim of this study is to develop a novel phantom for the evaluation of clinical CEST imaging settings, e.g., B0 and B1 field inhomogeneities, CEST contrast, and post-processing. We made a phantom composed of two slice sections: a grid section for local offset frequency evaluation and a sample section for CEST effect evaluation using different concentrations of an egg white albumin solution. On a 3 Tesla MR scanner, a phantom study was performed using CEST imaging; the mean B1 amplitudes were set at 1.2 and 1.9 µT, and CEST images with and without B0 corrections were acquired. Next, region of interest (ROI) analysis was performed for each slice. Then, CEST images with and without B0 corrections were compared at each B1 amplitude. The B0 corrected Z-spectrums at each local region in the grid section showed a shifting of the curve bottom to 0 ppm. Z-spectrum at B1 = 1.9 µT showed a broader curve shape than that at 1.2 µT. Moreover, MTRasym values at 3.5 ppm for each albumin sample at B1 = 1.9 µT were about two times higher than those at 1.2 µT. Our phantom enabled us to evaluate and optimize B0 inhomogeneity and the CEST effect at the B1 amplitude.


Asunto(s)
Albúminas , Imagen por Resonancia Magnética , Imagen por Resonancia Magnética/métodos , Fantasmas de Imagen
5.
Magn Reson Med ; 91(2): 716-734, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37749854

RESUMEN

PURPOSE: To evaluate the assumption in amide proton transfer weighted (APTw) imaging that the APT dominates over the relayed nuclear Overhauser enhancement (rNOE) and other CEST effects such as those from amines/guanidines, thereby providing imaging of mobile proteins/peptides. METHODS: We introduced two auxiliary asymmetric analysis metrics that can vary the relative contributions from amine/guanidinium CEST and other effects. By comparing these metrics with the conventional asymmetric analysis metric on healthy rat brains, we can approximately assess the contribution from amines/guanidines to APTw and determine whether the APT dominates over the rNOE effect. To further investigate the molecular origin of APTw, we used samples of dialyzed tissue homogenates to eliminate small metabolites and supernatants of homogenates to separate lipids from other components. RESULTS: When the APTw signal is positive using high saturation amplitudes (e.g., 2-3 µT), the contributions from amines/guanidines are significant and cannot be ignored. The APTw signal from the dialyzed homogenates and the controls has negligible changes, indicating that it primarily originates from macromolecules rather than small metabolites. Additionally, the APTw signals with low saturation amplitudes (e.g., 1 µT) were negative in tissue homogenates but positive in their supernatants, suggesting that proteins contribute positively to APTw signals, whereas lipids contribute negatively to it. CONCLUSION: The positive APTw signal using high saturation amplitudes could have significant contributions from soluble proteins through CEST, including amide/amine/guanidine proton transfer effects. In contrast, the negative APTw signal using low saturation amplitudes has significant contribution from lipids through rNOE.


Asunto(s)
Imagen por Resonancia Magnética , Protones , Ratas , Animales , Imagen por Resonancia Magnética/métodos , Amidas , Aminas , Guanidinas , Lípidos
6.
Quant Imaging Med Surg ; 13(12): 7879-7892, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38106293

RESUMEN

Background: When an ischemic stroke happens, it triggers a complex signalling cascade that may eventually lead to neuronal cell death if no reperfusion. Recently, the relayed nuclear Overhauser enhancement effect at -1.6 ppm [NOE(-1.6 ppm)] has been postulated may allow for a more in-depth analysis of the ischemic injury. This study assessed the potential utility of NOE(-1.6 ppm) in an ischemic stroke model. Methods: Diffusion-weighted imaging, perfusion-weighted imaging, and chemical exchange saturation transfer (CEST) magnetic resonance imaging (MRI) data were acquired from five rats that underwent scans at 9.4 T after middle cerebral artery occlusion. Results: The apparent diffusion coefficient (ADC), cerebral blood flow (CBF), and apparent exchange-dependent relaxations (AREX) at 3.5 ppm and NOE(-1.6 ppm) were quantified. AREX(3.5 ppm) and NOE(-1.6 ppm) were found to be hypointense and exhibited different signal patterns within the ischemic tissue. The NOE(-1.6 ppm) deficit areas were equal to or larger than the ADC deficit areas, but smaller than the AREX(3.5 ppm) deficit areas. This suggested that NOE(-1.6 ppm) might further delineate the acidotic tissue estimated using AREX(3.5 ppm). Since NOE(-1.6 ppm) is closely related to membrane phospholipids, NOE(-1.6 ppm) potentially highlighted at-risk tissue affected by lipid peroxidation and membrane damage. Altogether, the ADC/NOE(-1.6 ppm)/AREX(3.5 ppm)/CBF mismatches revealed four zones of increasing sizes within the ischemic tissue, potentially reflecting different pathophysiological information. Conclusions: Using CEST coupled with ADC and CBF, the ischemic tissue may thus potentially be separated into four zones to better understand the pathophysiology after stroke and improve ischemic tissue fate definition. Further verification of the potential utility of NOE(-1.6 ppm) may therefore lead to a more precise diagnosis.

7.
ArXiv ; 2023 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-37961738

RESUMEN

Purpose: Machine learning (ML) has been increasingly used to quantify chemical exchange saturation transfer (CEST) effect. ML models are typically trained using either measured data or fully simulated data. However, training with measured data often lacks sufficient training data, while training with fully simulated data may introduce bias due to limited simulations pools. This study introduces a new platform that combines simulated and measured components to generate partially synthetic CEST data, and to evaluate its feasibility for training ML models to predict amide proton transfer (APT) effect. Methods: Partially synthetic CEST signals were created using an inverse summation of APT effects from simulations and the other components from measurements. Training data were generated by varying APT simulation parameters and applying scaling factors to adjust the measured components, achieving a balance between simulation flexibility and fidelity. First, tissue-mimicking CEST signals along with ground truth information were created using multiple-pool model simulations to validate this method. Second, an ML model was trained individually on partially synthetic data, in vivo data, and fully simulated data, to predict APT effect in rat brains bearing 9L tumors. Results: Experiments on tissue-mimicking data suggest that the ML method using the partially synthetic data is accurate in predicting APT. In vivo experiments suggest that our method provides more accurate and robust prediction than the training using in vivo data and fully synthetic data. Conclusion: Partially synthetic CEST data can address the challenges in conventional ML methods.

8.
Magn Reson Med ; 90(6): 2400-2410, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37526017

RESUMEN

PURPOSE: To integrate 3D CEST EPI with an unevenly segmented RF irradiation module and preliminarily demonstrate it in the clinical setting. METHODS: A CEST MRI with unevenly segmented RF saturation was implemented, including a long primary RF saturation to induce the steady-state CEST effect, maintained with repetitive short secondary RF irradiation between readouts. This configuration reduces relaxation-induced blur artifacts during acquisition, allowing fast 3D spatial coverage. Numerical simulations were performed to select parameters such as flip angle (FA), short RF saturation duration (Ts2), and the number of readout segments. The sequence was validated experimentally with data from a phantom, healthy volunteers, and a brain tumor patient. RESULTS: Based on the numerical simulation and l-carnosine gel phantom experiment, FA, Ts2, and the number of segments were set to 20°, 0.3 s, and the range from 4 to 8, respectively. The proposed method minimized signal modulation in the human brain images in the kz direction during the acquisition and provided the blur artifacts-free CEST contrast over the whole volume. Additionally, the CEST contrast in the tumor tissue region is higher than in the contralateral normal tissue region. CONCLUSIONS: It is feasible to implement a highly accelerated 3D EPI CEST imaging with unevenly segmented RF irradiation.


Asunto(s)
Neoplasias Encefálicas , Imagen por Resonancia Magnética , Humanos , Estudios de Factibilidad , Imagen por Resonancia Magnética/métodos , Neoplasias Encefálicas/diagnóstico por imagen , Encéfalo/diagnóstico por imagen , Fantasmas de Imagen
9.
Magn Reson Imaging ; 102: 201-202, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37353181

RESUMEN

The CEST2022 workshop was held at the Emory Conference Center Hotel from August 7th to 10th, 2022, and attracted over a hundred international participants from North America, Europe, and Asia. The workshop consisted of four plenary talks, 10 scientific sessions, nine invited talks, and 40 talks selected from abstracts. Four discussion sessions were also conducted to build consensus on CEST imaging standardization and quantification. We thank Professors Peter van Zijl, Ravinder Reddy, and Dean Sherry for their Sunday afternoon education session and Professors Linda Knutsson, John Gore, Mortiz Zaiss, and Fahmeed Hyder for their plenary lectures. Abstracts selected for oral presentations were invited to submit to Magnetic Resonance Imaging for peer review and are included in this special issue. Here, we present a summary of the articles featured in this issue.


Asunto(s)
Imagen por Resonancia Magnética , Protones , Humanos , Imagen por Resonancia Magnética/métodos
10.
Magn Reson Med ; 90(5): 1958-1968, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37335834

RESUMEN

PURPOSE: CEST MRI detects complex tissue changes following acute stroke. Our study aimed to test if spinlock model-based fitting of the quasi-steady-state (QUASS)-reconstructed equilibrium CEST MRI improves the determination of multi-pool signal changes over the commonly-used model-free Lorentzian fitting in acute stroke. THEORY AND METHODS: Multiple three-pool CEST Z-spectra were simulated using Bloch-McConnell equations for a range of T1 , relaxation delay, and saturation times. The multi-pool CEST signals were solved from the simulated Z-spectra to test the accuracy of routine Lorentzian (model-free) and spinlock (model-based) fittings without and with QUASS reconstruction. In addition, multiparametric MRI scans were obtained in rat models of acute stroke, including relaxation, diffusion, and CEST Z-spectrum. Finally, we compared model-free and model-based per-pixel CEST quantification in vivo. RESULTS: The spinlock model-based fitting of QUASS CEST MRI provided a nearly T1 -independent determination of multi-pool CEST signals, advantageous over the fittings of apparent CEST MRI (model-free and model-based). In vivo data also demonstrated that the spinlock model-based QUASS fitting captured significantly different changes in semisolid magnetization transfer (-0.9 ± 0.8 vs. 0.3 ± 0.8%), amide (-1.1 ± 0.4 vs. -0.5 ± 0.2%), and guanidyl (1.0 ± 0.4 vs. 0.7 ± 0.3%) signals over the model-free Lorentzian analysis. CONCLUSION: Our study demonstrated that spinlock model-based fitting of QUASS CEST MRI improved the determination of the underlying tissue changes following acute stroke, promising further clinical translation of quantitative CEST imaging.


Asunto(s)
Imágenes de Resonancia Magnética Multiparamétrica , Accidente Cerebrovascular , Ratas , Animales , Accidente Cerebrovascular/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Amidas
11.
Magn Reson Med ; 90(4): 1502-1517, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37317709

RESUMEN

PURPOSE: Accurately quantifying the amide proton transfer (APT) effect and the underlying exchange parameters is crucial for its applications, but previous studies have reported conflicting results. In these quantifications, the CEST effect from the fast exchange amine was always ignored because it was considered weak with low saturation powers. This paper aims to evaluate the influence of the fast exchange amine CEST on the quantification of APT at low saturation powers. METHODS: A quantification method with low and high saturation powers was used to distinguish APT from the fast exchange amine CEST effect. Simulations were conducted to assess the method's capability to separate APT from the fast exchange amine CEST effect. Animal experiments were performed to assess the relative contributions from the fast exchange amine and amide to CEST signals at 3.5 ppm. Three APT quantification methods, each with varying degrees of contamination from the fast exchange amine, were employed to process the animal data to assess the influence of the amine on the quantification of APT effect and the exchange parameters. RESULTS: The relative size of the fast exchange amine CEST effect to APT effect gradually increases with increasing saturation power. At 9.4 T, it increases from approximately 20% to 40% of APT effect with a saturation power increase from 0.25 to 1 µT. CONCLUSION: The fast exchange amine CEST effect leads overestimation of APT effect, fitted amide concentration, and amide-water exchange rate, potentially contributing to the conflicting results reported in previous studies.


Asunto(s)
Imagen por Resonancia Magnética , Protones , Animales , Imagen por Resonancia Magnética/métodos , Amidas , Aminas , Agua
12.
Quant Imaging Med Surg ; 13(5): 3001-3012, 2023 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-37179906

RESUMEN

Background: The diagnosis and treatment of childhood neuroblastoma (NB) varies with different risk groups, thus requiring accurate preoperative risk assessment. This study aimed to verify the feasibility of amide proton transfer (APT) imaging in risk stratification of abdominal NB in children, and compare it with the serum neuron-specific enolase (NSE). Methods: This prospective study enrolled 86 consecutive pediatric volunteers with suspected NB, and all subjects underwent abdominal APT imaging on a 3T magnetic resonance imaging scanner. A 4-pool Lorentzian fitting model was used to mitigate motion artifacts and separate the APT signal from the contaminating ones. The APT values were measured from tumor regions delineated by two experienced radiologists. The one-way analysis of variance, independent-sample t-test, Mann-Whitney U-test, and receiver operating characteristic analysis were performed to evaluate and compare the risk stratification performance of the APT value and serum NSE index-a routine biomarker of NB in clinics. Results: Thirty-four cases (mean age, 38.6±32.4 months; 5 very-low-risk, 5 low-risk, 8 intermediate-risk and 16 high-risk ones) were included in the final analysis. The APT values were significantly higher in high-risk NB (5.80%±1.27%) than in the non-high-risk group (3.88%±1.01%) composed of the other three risk groups (P<0.001). However, there was no significant difference (P=0.18) in NSE levels between the high-risk (93.05±97.14 ng/mL) and non-high-risk groups (41.45±30.99 ng/mL). The associated area under the curve (AUC) of the APT parameter (AUC =0.89) in differentiating high-risk NB from non-high-risk NB was significantly higher (P=0.03) than that of NSE (AUC =0.64). Conclusions: As an emerging non-invasive magnetic resonance imaging technique, APT imaging has a promising prospect for distinguishing high-risk NB from non-high-risk NB in routine clinical applications.

13.
Magn Reson Med ; 90(2): 722-736, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37052377

RESUMEN

PURPOSE: To propose a novel Numerical fitting method of the Extrapolated semisolid Magnetization transfer Reference (NEMR) signal for quantifying the CEST effect. THEORY AND METHODS: Modified two-pool Bloch-McConnell equations were used to numerically fit the magnetization transfer (MT) and direct water saturation (DS) signals at far off-resonance frequencies, which was subsequently extrapolated into the frequency range of amide proton transfer (APT) and nuclear Overhauser enhancement (NOE) pools. Then the subtraction of the fitted two-pool z-spectrum and the experimentally acquired z-spectrum yielded APT# and NOE# signals mostly free of MT and DS contamination. Several strategies were used to accelerate the NEMR fitting. Furthermore, the proposed NEMR method was compared with the conventional extrapolated semisolid magnetization transfer reference (EMR) and magnetization transfer ratio asymmetry (MTRasym ) methods in simulations and stroke patients. RESULTS: The combination of RF downsampling, MT lineshape look-up table, and conversion of MATLAB code to C code accelerated the NEMR fitting by over 2700-fold. Monte-Carlo simulations showed that NEMR had higher accuracy than EMR and eliminated the requirement of the steady-state condition. In ischemic stroke patients, the NEMR maps at 1 µT removed hypointense artifacts seen on EMR and MTRasym images, and better depicted stroke lesions than EMR. For NEMR, NOE# yielded significantly (p < 0.05) stronger signal contrast between stroke and normal tissues than APT# at 1 µT. CONCLUSION: The proposed NEMR method is suitable for arbitrary saturation settings and can remove MT and DS contamination from the CEST signal for improved detection of ischemic stroke.


Asunto(s)
Neoplasias Encefálicas , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Humanos , Neoplasias Encefálicas/patología , Imagen por Resonancia Magnética/métodos , Algoritmos , Accidente Cerebrovascular/diagnóstico por imagen , Protones , Amidas
14.
Magn Reson Imaging ; 99: 123-129, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36822450

RESUMEN

PURPOSE: Amide proton transfer-weighted (APTw) imaging was an effective tool to reveal the tissue acidosis of acute ischemic stroke. This study aimed to evaluate the ability of APTw MRI to distinguish progressive penumbra and benign oligemia in the diffusion-perfusion mismatch region. MATERIALS AND METHODS: 38 acute cerebral infarction patients who underwent a comprehensive MRI examination, including diffusion-weighted imaging (DWI), perfusion-weighted imaging (PWI), APT imaging, and a follow-up scan in one week were recruited. There were 12 DWI/PWI match cases. The DWI/PWI mismatch patients were divided into 10 progressive cases and 16 stable cases according to the lesion size on the follow-up DWI image compared to the admission scan. Three ROIs: infarction lesion, peripheral, and contralateral normal regions were measured on each subject's MTRasym map. The Friedman test was used to compare the changes of MTRasym among three different regions within each group. The Kruskal-Wallis ANOVA test was used to compare them among the same region of different groups. The correlation between the MTRasym of the peripheral region and the lesion enlargement was analyzed by the Spearman test. RESULTS: The MTRasym at the infarction lesion of all three groups showed significant decrease to the contralateral normal tissue. In the progressive mismatch group, the MTRasym at the peripheral region within the DWI/PWI mismatch showed a significant difference with the contralateral normal region and no difference with the infarct core. Whereas both the MTRasym at the peripheral region of the stable mismatch and match groups had no significant difference with the contralateral side, but the differences were significant from those of the central core. When comparing the peripheral region of three groups, the MTRasym of the progressive mismatch group showed a significant decrease to that of the stable mismatch and match groups. The MTRasym of the peripheral region showed a negative correlation with lesion enlargement. CONCLUSION: APTw imaging is promising to stratify the heterogeneous PWI/DWI mismatch region and benefit the clinical treatment.


Asunto(s)
Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Humanos , Protones , Amidas , Accidente Cerebrovascular/patología , Imagen por Resonancia Magnética/métodos , Imagen de Difusión por Resonancia Magnética/métodos , Perfusión , Infarto
15.
Quant Imaging Med Surg ; 13(2): 812-824, 2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36819246

RESUMEN

Background: More effective risk stratification of prostate cancer (PCa) than that possible with current methods can reduce undertreatment and guard against overtreatment. The aim of this study is to validate the differences and combined effects of amide proton transfer (APT) imaging and apparent diffusion coefficient (ADC) in discriminating the PCa grade group (GG) ≤2 from GG ≥3 PCa. Methods: This is an ongoing prospective study conducted in the radiology department of Nanxishan Hospital of Guangxi Zhuang Autonomous Region. Patients pathologically diagnosed with PCa were enrolled consecutively according to the eligibility criteria. A total of 180 patients (age range, 42-92 years) were included in this study. Using histopathology as the reference standard, we placed 71 cases in GG ≤2 (mean age 67.03±8.696 years) and 109 cases in GG ≥3 (age 69.60±9.638 years). Magnetic resonance imaging (MRI) parameters, including APT and ADC values, were analyzed using an independent samples t-test and binary logistic regression analysis stratified with GG. Receiver operating characteristic curve was used to analyze the diagnostic performance for different parameters distinguishing GG ≤2 and GG ≥3. Results: APT [odds ratio (OR) for the transitional zone (TZ) PCa: 3.20, 95% CI: 1.14-8.98, P=0.02; OR for the peripheral zone (PZ) PCa: 86.32, 95% CI: 13.24-562.88, P=0.003] and ADC values (OR for TZ PCa: 89.79; 95% CI: 2.85-2,827.99, P=0.01; OR for PZ PCa: 39.92; 95% CI: 3.22-494.18, P=0.004) were independent predictors that differentiated the GG of patients. The sensitivity and specificity of the APT values were 61.1% and 81.0%, respectively, while the sensitivity and specificity of the ADC values were 83.3% and 61.9%, respectively. The optimal cutoff value of APT was 3.35% and which of ADC was 1.25×10-3 mm2/s in TZ origin PCa. At the optimal cutoff values of 3.31% (APT) and 0.79×10-3 mm2/s (ADC) in PZ PCa, the sensitivity and specificity of the APT values were 74.0% and 83.6%, respectively, while the sensitivity and specificity of the ADC values were 94.0% and 53.4%, respectively. The area under the curve of the combination of APT and ADC was significantly higher than either of APT or ADC alone in Delong test (TZ: P=0.002 and P=0.020; PZ: P=0.033 and P<0.001). Conclusions: APT and ADC have complementary effects on the sensitivity and specificity for identifying different PCa GGs. A combination model of APT and ADC could improve the diagnostic efficacy of PCa differentiation.

16.
Cancer Imaging ; 23(1): 3, 2023 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-36611191

RESUMEN

BACKGROUND: Early diagnosis of prostate cancer improves its prognosis, while it is essential to upgrade screening tools. This study aimed to explore the value of a novel functional magnetic resonance imaging (MRI) technique, namely amide proton transfer (APT)-weighted MRI, combined with serum prostate-specific antigen (PSA) levels to differentiate malignant prostate lesions from benign prostate lesions. METHODS: Data of patients who underwent prostate examinations at Chongqing University Cancer Hospital between July 2019 and March 2022 were retrospectively analyzed. All patients underwent T2-weighted imaging (T2WI), APT, diffusion-weighted imaging (DWI), and dynamic contrast-enhanced (DCE) MRI. Two radiologists analyzed the images independently. The ability of the quantitative parameters alone or in different combinations in differentiating malignant prostate lesions from benign prostate lesions were compared by using receiver operating characteristic (ROC) curves. According to the DeLong test, the combined parameters were significantly different from the corresponding single parameter (P < 0.05). RESULTS: A total of 79 patients were finally enrolled, including 52 patients in the malignant group and 27 patients in the benign group. The separate assessment of indexes revealed that APTmax, APTmean, mean apparent diffusion coefficient (ADCmean), ADCmax, ADCmin, tPAD, free prostate-specific antigen (FPSA), FPSA/total prostate-specific antigen (tPSA), and PSA density (PSAD) were significantly different between the two groups (P < 0.05), while APTmin was not significantly different between the two groups (P > 0.05). APTmax and APTmean had the high values of area under the ROC curve (AUC), which were 0.780 and 0.710, respectively. APTmax had a high sensitivity, and APTmean had a high specificity. The combination of APTmax, APTmean, ADCmean, and PSAD had the highest AUC value (AUC: 0.880, sensitivity: 86.540, specificity: 78.260). CONCLUSION: APTmax, APTmean, ADCmean, ADCmin, tPAD, FPSA, and PSAD showed to have a high value in differentiating malignant prostate lesions from benign prostate lesions in the separate assessment of indexes. The combination of APTmax, APTmean, ADCmean, and PSAD had the highest diagnostic value.


Asunto(s)
Antígeno Prostático Específico , Próstata , Masculino , Humanos , Estudios Retrospectivos , Protones , Imagen por Resonancia Magnética/métodos , Curva ROC , Imagen de Difusión por Resonancia Magnética/métodos , Amidas , Sensibilidad y Especificidad
17.
Front Oncol ; 12: 939260, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36483050

RESUMEN

Many drugs that show potential in animal models of glioblastoma (GBM) fail to translate to the clinic, contributing to a paucity of new therapeutic options. In addition, animal model development often includes histologic assessment, but multiparametric/multimodality imaging is rarely included despite increasing utilization in patient cancer management. This study developed an intracranial recurrent, drug-resistant, human-derived glioblastoma tumor in Sprague-Dawley Rag2-Rag2 tm1Hera knockout rat and was characterized both histologically and using multiparametric/multimodality neuroimaging. Hybrid 18F-fluoroethyltyrosine positron emission tomography and magnetic resonance imaging, including chemical exchange saturation transfer (18F-FET PET/CEST MRI), was performed for full tumor viability determination and characterization. Histological analysis demonstrated human-like GBM features of the intracranially implanted tumor, with rapid tumor cell proliferation (Ki67 positivity: 30.5 ± 7.8%) and neovascular heterogeneity (von Willebrand factor VIII:1.8 to 5.0% positivity). Early serial MRI followed by simultaneous 18F-FET PET/CEST MRI demonstrated consistent, predictable tumor growth, with exponential tumor growth most evident between days 35 and 49 post-implantation. In a second, larger cohort of rats, 18F-FET PET/CEST MRI was performed in mature tumors (day 49 post-implantation) for biomarker determination, followed by evaluation of single and combination therapy as part of the model development and validation. The mean percentage of the injected dose per mL of 18F-FET PET correlated with the mean %CEST (r = 0.67, P < 0.05), but there was also a qualitative difference in hot spot location within the tumor, indicating complementary information regarding the tumor cell demand for amino acids and tumor intracellular mobile phase protein levels. Finally, the use of this glioblastoma animal model for therapy assessment was validated by its increased overall survival after treatment with combination therapy (temozolomide and idasanutlin) (P < 0.001). Our findings hold promise for a more accurate tumor viability determination and novel therapy assessment in vivo in a recently developed, reproducible, intracranial, PDX GBM.

18.
Magn Reson Med ; 88(6): 2633-2644, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36178234

RESUMEN

PURPOSE: Chemical exchange saturation transfer (CEST) imaging measurement depends not only on the labile proton concentration and pH-dependent exchange rate but also on experimental conditions, including the relaxation delay and radiofrequency (RF) saturation time. Our study aimed to extend a quasi-steady-state (QUASS) solution to a modified multi-slice CEST MRI sequence and test if it provides enhanced pH imaging after acute stroke. METHODS: Our study derived the QUASS solution for a modified multislice CEST MRI sequence with an unevenly segmented RF saturation between image readout and signal averaging. Numerical simulation was performed to test if the generalized QUASS solution corrects the impact of insufficiently long relaxation delay, primary and secondary saturation times, and multi-slice readout. In addition, multiparametric MRI scans were obtained after middle cerebral artery occlusion, including relaxation and CEST Z-spectrum, to evaluate the performance of QUASS CEST MRI in a rodent acute stroke model. We also performed Lorentzian fitting to isolate multi-pool CEST contributions. RESULTS: The QUASS analysis enhanced pH-weighted magnetization transfer asymmetry contrast over the routine apparent CEST measurements in both contralateral normal (-3.46% ± 0.62% (apparent) vs. -3.67% ± 0.66% (QUASS), P < 0.05) and ischemic tissue (-5.53% ± 0.68% (apparent) vs. -5.94% ± 0.73% (QUASS), P < 0.05). Lorentzian fitting also showed significant differences between routine and QUASS analysis of ischemia-induced changes in magnetization transfer, amide, amine, guanidyl CEST, and nuclear Overhauser enhancement (-1.6 parts per million) effects. CONCLUSION: Our study demonstrated that generalized QUASS analysis enhanced pH MRI contrast and improved quantification of the underlying CEST contrast mechanism, promising for further in vivo applications.


Asunto(s)
Protones , Accidente Cerebrovascular , Algoritmos , Amidas , Aminas , Dimaprit/análogos & derivados , Humanos , Concentración de Iones de Hidrógeno , Interpretación de Imagen Asistida por Computador/métodos , Imagen por Resonancia Magnética/métodos , Accidente Cerebrovascular/diagnóstico por imagen
19.
Front Oncol ; 12: 852076, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35978813

RESUMEN

Background: Amide proton transfer (APT) imaging as an emerging MRI approach has been used for distinguishing tumor recurrence (TR) and treatment effects (TEs) in glioma patients, but the initial results from recent studies are different. Aim: The aim of this study is to systematically review and quantify the diagnostic performance of APT in assessing treatment response in patients with post-treatment gliomas. Methods: A systematic search in PubMed, EMBASE, and the Web of Science was performed to retrieve related original studies. For the single and added value of APT imaging in distinguishing TR from TEs, we calculated pooled sensitivity and specificity by using Bayesian bivariate meta-analyses. Results: Six studies were included, five of which reported on single APT imaging parameters and four of which reported on multiparametric MRI combined with APT imaging parameters. For single APT imaging parameters, the pooled sensitivity and specificity were 0.85 (95% CI: 0.75-0.92) and 0.88 (95% CI: 0.74-0.97). For multiparametric MRI including APT, the pooled sensitivity and specificity were 0.92 (95% CI: 0.85-0.97) and 0.83 (95% CI: 0.55-0.97), respectively. In addition, in the three studies reported on both single and added value of APT imaging parameters, the combined imaging parameters further improved diagnostic performance, yielding pooled sensitivity and specificity of 0.91 (95% CI: 0.80-0.97) and 0.92 (95% CI: 0.79-0.98), respectively, but the pooled sensitivity was 0.81 (95% CI: 0.65-0.93) and specificity was 0.82 (95% CI: 0.61-0.94) for single APT imaging parameters. Conclusion: APT imaging showed high diagnostic performance in assessing treatment response in patients with post-treatment gliomas, and the addition of APT imaging to other advanced MRI techniques can improve the diagnostic accuracy for distinguishing TR from TE.

20.
Tomography ; 8(4): 1974-1986, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-36006063

RESUMEN

Amide proton transfer (APT)-weighted MRI is a promising molecular imaging technique that has been employed in clinic for detection and grading of brain tumors. MTRasym, the quantification method of APT, is easily influenced by B0 inhomogeneity and causes artifacts. Current model-free interpolation methods have enabled moderate B0 correction for middle offsets, but have performed poorly at limbic offsets. To address this shortcoming, we proposed a practical B0 correction approach that is suitable under time-limited sparse acquisition scenarios and for B1 ≥ 1 µT under 3T. In this study, this approach employed a simplified Lorentzian model containing only two pools of symmetric water and asymmetric solutes, to describe the Z-spectral shape with wide and 'invisible' CEST peaks. The B0 correction was then performed on the basis of the fitted two-pool Lorentzian lines, instead of using conventional model-free interpolation. The approach was firstly evaluated on densely sampled Z-spectra data by using the spline interpolation of all acquired 16 offsets as the gold standard. When only six offsets were available for B0 correction, our method outperformed conventional methods. In particular, the errors at limbic offsets were significantly reduced (n = 8, p < 0.01). Secondly, our method was assessed on the six-offset APT data of nine brain tumor patients. Our MTRasym (3.5 ppm), using the two-pool model, displayed a similar contrast to the vendor-provided B0-orrected MTRasym (3.5 ppm). While the vendor failed in correcting B0 at 4.3 and 2.7 ppm for a large portion of voxels, our method enabled well differentiation of B0 artifacts from tumors. In conclusion, the proposed approach could alleviate analysis errors caused by B0 inhomogeneity, which is useful for facilitating the comprehensive metabolic analysis of brain tumors.


Asunto(s)
Neoplasias Encefálicas , Protones , Amidas , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/patología , Dimaprit/análogos & derivados , Humanos , Imagen por Resonancia Magnética/métodos , Agua
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA