Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
NMR Biomed ; 32(7): e4097, 2019 07.
Article in English | MEDLINE | ID: mdl-31058381

ABSTRACT

Although magnetization transfer (MT) has been widely used in brain MRI, for example in brain inflammation and multiple sclerosis, the detailed molecular origin of MT effects and the role that proteins play in MT remain unclear. In this work, a proteoliposome model system was used to mimic the myelin environment and to examine the roles of protein, cholesterol, brain cerebrosides, and sphingomyelin embedded in the liposome matrix. Exchange parameters were determined using a double-quantum filter experiment. The goal was to determine the relative contributions to exchange and MT of cerebrosides, sphingomyelin, cholesterol, and proteins in 1,2-dimyristoyl-sn-glycero-3-phosphocholine bilayers. The main finding was that cerebrosides produced the strongest exchange effects, and that these were even more pronounced than those found for proteins. Sphingomyelin (which also has exchangeable groups at the head of the fatty acid chains, albeit closer to the lipid acyl chains) and cholesterol showed only minimal transfer. Overall, the extracted exchange rates appeared much smaller than commonly assumed for -OH and -NH groups.


Subject(s)
Liposomes/chemistry , Magnetic Resonance Spectroscopy , Myelin Sheath/chemistry , Proteolipids/chemistry , Lipids/chemistry , Water/chemistry
2.
Eur Radiol ; 29(9): 4957-4967, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30809720

ABSTRACT

OBJECTIVES: The purpose of this study was to investigate the association of relaxation-compensated chemical exchange saturation transfer (CEST) MRI with overall survival (OS) and progression-free survival (PFS) in newly diagnosed high-grade glioma (HGG) patients. METHODS: Twenty-six patients with newly diagnosed high-grade glioma (WHO grades III-IV) were included in this prospective IRB-approved study. CEST MRI was performed on a 7.0-T whole-body scanner. Association of patient OS/PFS with relaxation-compensated CEST MRI (amide proton transfer (APT), relayed nuclear Overhauser effect (rNOE)/NOE, downfield-rNOE-suppressed APT (dns-APT)) and diffusion-weighted imaging (apparent diffusion coefficient) were assessed using the univariate Cox proportional hazards regression model. Hazard ratios (HRs) and corresponding 95% confidence intervals were calculated. Furthermore, OS/PFS association with clinical parameters (age, gender, O6-methylguanine-DNA methyltransferase (MGMT) promotor methylation status, and therapy: biopsy + radio-chemotherapy vs. debulking surgery + radio-chemotherapy) were tested accordingly. RESULTS: Relaxation-compensated APT MRI was significantly correlated with patient OS (HR = 3.15, p = 0.02) and PFS (HR = 1.83, p = 0.009). The strongest association with PFS was found for the dns-APT metric (HR = 2.61, p = 0.002). These results still stand for the relaxation-compensated APT contrasts in a homogenous subcohort of n = 22 glioblastoma patients with isocitrate dehydrogenase (IDH) wild-type status. Among the tested clinical parameters, patient age (HR = 1.1, p = 0.001) and therapy (HR = 3.68, p = 0.026) were significant for OS; age additionally for PFS (HR = 1.04, p = 0.048). CONCLUSION: Relaxation-compensated APT MRI signal intensity is associated with overall survival and progression-free survival in newly diagnosed, previously untreated glioma patients and may, therefore, help to customize treatment and response monitoring in the future. KEY POINTS: • Amide proton transfer (APT) MRI signal intensity is associated with overall survival and progression in glioma patients. • Relaxation compensation enhances the information value of APT MRI in tumors. • Chemical exchange saturation transfer (CEST) MRI may serve as a non-invasive biomarker to predict prognosis and customize treatment.


Subject(s)
Brain Neoplasms/diagnostic imaging , Diffusion Magnetic Resonance Imaging/methods , Glioma/diagnostic imaging , Adult , Aged , Amides , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Disease Progression , Female , Glioblastoma/diagnostic imaging , Glioblastoma/enzymology , Glioblastoma/pathology , Glioma/enzymology , Glioma/pathology , Humans , Isocitrate Dehydrogenase/metabolism , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Grading , Prognosis , Progression-Free Survival , Prospective Studies , Protons
3.
Magn Reson Med ; 81(1): 573-582, 2019 01.
Article in English | MEDLINE | ID: mdl-29851141

ABSTRACT

PURPOSE: To investigate the effect of a frequency drift of the static magnetic field on 3D CEST MRI based on glycosaminoglycans (GAGs) of articular cartilage at 7 T and to introduce a retrospective correction method that uses the phase images of the gradient-echo readout. METHODS: Repeated gagCEST and B0 measurements were performed in a glucose model solution and in vivo in the knee joint of 3 healthy volunteers at 7 T. Phase images of the modified 3D rectangular spiral centric-reordered gradient-echo CEST sequence were used to quantify and compensate the apparent frequency drift in repeated gagCEST measurements. RESULTS: The frequency drift of the MRI scanner strongly influences the gagCEST signal in the articular cartilage of the human knee joint. The gagCEST signal in the articular cartilage is changed by 0.18%/Hz while an average drift of 0.7 ± 0.2 Hz/minute was observed. The proposed correction method can be applied retrospectively without the need of additional measurements and provides improved comparability and reproducibility for gagCEST studies. This correction method may also be of interest for other applications of CEST MRI. CONCLUSION: Prospective or retrospective correction of the frequency drift of the MRI scanner is essential for reproducible gagCEST measurements. The proposed retrospective correction method fulfills this requirement without the need of additional measurements.


Subject(s)
Cartilage, Articular/diagnostic imaging , Glycosaminoglycans/chemistry , Magnetic Fields , Magnetic Resonance Imaging , Adult , Algorithms , Blood Glucose/analysis , Contrast Media , Female , Healthy Volunteers , Humans , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Knee Joint/diagnostic imaging , Male , Phantoms, Imaging , Protons , Reproducibility of Results , User-Computer Interface
4.
J Magn Reson Imaging ; 49(3): 777-785, 2019 03.
Article in English | MEDLINE | ID: mdl-30133046

ABSTRACT

BACKGROUND: Chemical exchange saturation transfer (CEST) is a novel MRI technique applied to brain tumor patients. PURPOSE: To investigate the anatomic location dependence of CEST MRI obtained at 7T and histopathological/molecular parameters in WHO IV° glioma patients. STUDY TYPE: Analytic prospective study. POPULATION: Twenty-one patients with newly diagnosed WHO IV° gliomas were studied prior to surgery; 11 healthy volunteers were investigated. FIELD STRENGTH/SEQUENCE: Conventional MRI (contrast-enhanced, T2 w and diffusion-weighted imaging) at 3T and T2 w and CEST MRI at 7T was performed for patients and both patients and volunteers. ASSESSMENT: Mean CEST signal intensities (nuclear-Overhauser-enhancement [NOE], amide-proton-transfer [APT], downfield NOE-suppressed APT [dns-APT]), ADC values, and histopathological/molecular parameters were evaluated with regard to hemisphere location and contact with the subventricular zone. CEST signal intensities of cerebral tissue of healthy volunteers were evaluated with regard to hemisphere discrimination. STATISTICAL TESTS: Spearman correlation, Mann-Whitney U-test, Wilcoxon signed-rank-test, Fisher's exact test, and area under the receiver operating curve. RESULTS: Maximum APT and dns-APT signal intensities were significantly different in right vs. left hemisphere gliomas (P = 0.037 and P = 0.007), but not in right vs. left hemisphere cerebral tissue of healthy subjects (P = 0.062-0.859). Mean ADC values were significantly decreased in right vs. left hemisphere gliomas (P = 0.044). Mean NOE signal intensity did not differ significantly between gliomas of either hemisphere (P = 0.820), but in case of subventricular zone contact (P = 0.047). A significant correlation was observed between APT and dns-APT and ADC signal intensities (rs = -0.627, P = 0.004 and rs = -0.534, P = 0.019), but not between NOE and ADC (rs = -0.341, P = 0.154). Histopathological/molecular parameters were not significantly different concerning the tumor location (P = 0.104-1.000, P = 0.286-0.696). DATA CONCLUSION: APT, dns-APT, and ADC were inversely correlated and depended on the gliomas' hemisphere location. NOE showed significant dependence on subventricular zone contact. Location dependency of APT- and NOE-mediated CEST effects should be considered in clinical investigations of CEST MRI. LEVEL OF EVIDENCE: 2 Technical Efficacy: Stage 3 J. Magn. Reson. Imaging 2019;49:777-785.


Subject(s)
Brain Neoplasms/diagnostic imaging , Brain/diagnostic imaging , Diffusion Magnetic Resonance Imaging , Glioblastoma/diagnostic imaging , Gliosarcoma/diagnostic imaging , Adult , Healthy Volunteers , Humans , Magnetic Resonance Imaging , Middle Aged , Prospective Studies , Reproducibility of Results , Young Adult
5.
Oncotarget ; 9(47): 28772-28783, 2018 Jun 19.
Article in English | MEDLINE | ID: mdl-29983895

ABSTRACT

PURPOSE: To prospectively investigate chemical exchange saturation transfer (CEST) MRI in glioblastoma patients as predictor of early tumor progression after first-line treatment. EXPERIMENTAL DESIGN: Twenty previously untreated glioblastoma patients underwent CEST MRI employing a 7T whole-body scanner. Nuclear Overhauser effect (NOE) as well as amide proton transfer (APT) CEST signals were isolated using Lorentzian difference (LD) analysis and relaxation compensated by the apparent exchange-dependent relaxation rate (AREX) evaluation. Additionally, NOE-weighted asymmetric magnetic transfer ratio (MTRasym) and downfield-NOE-suppressed APT (dns-APT) were calculated. Patient response to consecutive treatment was determined according to the RANO criteria. Mean signal intensities of each contrast in the whole tumor area were compared between early-progressive and stable disease. RESULTS: Pre-treatment tumor signal intensity differed significantly regarding responsiveness to first-line therapy in NOE-LD (p = 0.0001), NOE-weighted MTRasym (p = 0.0186) and dns-APT (p = 0.0328) contrasts. Hence, significant prediction of early progression was possible employing NOE-LD (AUC = 0.98, p = 0.0005), NOE-weighted MTRasym (AUC = 0.83, p = 0.0166) and dns-APT (AUC = 0.80, p = 0.0318). The NOE-LD provided the highest sensitivity (91%) and specificity (100%). CONCLUSIONS: CEST derived contrasts, particularly NOE-weighted imaging and dns-APT, yielded significant predictors of early progression after fist-line therapy in glioblastoma. Therefore, CEST MRI might be considered as non-invasive tool for customization of treatment in the future.

6.
Neuro Oncol ; 20(12): 1661-1671, 2018 11 12.
Article in English | MEDLINE | ID: mdl-29733378

ABSTRACT

Background: Early identification of prognostic superior characteristics in glioma patients such as isocitrate dehydrogenase (IDH) mutation and O6-methylguanine-DNA-methyltransferase (MGMT) promoter methylation status is of great clinical importance. The study purpose was to investigate the non-invasive predictability of IDH mutation status, MGMT promoter methylation, and differentiation of low-grade versus high-grade glioma (LGG vs HGG) in newly diagnosed patients employing relaxation-compensated multipool chemical exchange saturation transfer (CEST) MRI at 7.0 Tesla. Methods: Thirty-one patients with newly diagnosed glioma were included in this prospective study. CEST MRI was performed at a 7T whole-body scanner. Nuclear Overhauser effect (NOE) and isolated amide proton transfer (APT; downfield NOE-suppressed APT = dns-APT) CEST signals (mean value and 90th signal percentile) were quantitatively investigated in the whole tumor area with regard to predictability of IDH mutation, MGMT promoter methylation status, and differentiation of LGG versus HGG. Statistics were performed using receiver operating characteristic (ROC) and area under the curve (AUC) analysis. Results were compared with advanced MRI methods (apparent diffusion coefficient and relative cerebral blood volume ROC/AUC analysis) obtained at 3T. Results: dns-APT CEST yielded highest AUCs in IDH mutation status prediction (dns-APTmean = 91.84%, P < 0.01; dns-APT90 = 97.96%, P < 0.001). Furthermore, dns-APT metrics enabled significant differentiation of LGG versus HGG (AUC: dns-APTmean = 0.78, P < 0.05; dns-APT90 = 0.83, P < 0.05). There was no significant difference regarding MGMT promoter methylation status at any contrast (P > 0.05). Conclusions: Relaxation-compensated multipool CEST MRI, particularly dns-APT imaging, enabled prediction of IDH mutation status and differentiation of LGG versus HGG and should therefore be considered as a non-invasive MR biomarker in the diagnostic workup.


Subject(s)
Brain Neoplasms/genetics , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Diffusion Magnetic Resonance Imaging/methods , Glioma/genetics , Isocitrate Dehydrogenase/genetics , Mutation , Tumor Suppressor Proteins/genetics , Adult , Aged , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Female , Follow-Up Studies , Glioma/diagnostic imaging , Glioma/pathology , Humans , Male , Middle Aged , Neoplasm Grading , Predictive Value of Tests , Prospective Studies , ROC Curve
7.
NMR Biomed ; 31(6): e3920, 2018 06.
Article in English | MEDLINE | ID: mdl-29672976

ABSTRACT

A novel MRI contrast is proposed which enables the selective detection of endogenous bulk mobile proteins in vivo. Such a non-invasive imaging technique may be of particular interest for many diseases associated with pathological alterations of protein expression, such as cancer and neurodegenerative disorders. Specificity to mobile proteins was achieved by the selective measurement of intramolecular spin diffusion and the removal of semi-solid macromolecular signal components by a correction procedure. For this purpose, the approach of chemical exchange saturation transfer (CEST) was extended to a radiofrequency (RF) irradiation scheme at two different frequency offsets (dualCEST). Using protein model solutions, it was demonstrated that the dualCEST technique allows the calculation of an image contrast which is exclusively sensitive to changes in concentration, molecular size and the folding state of mobile proteins. With respect to application in humans, dualCEST overcomes the selectivity limitations at relatively low magnetic field strengths, and thus enables examinations on clinical MR scanners. The feasibility of dualCEST examinations in humans was verified by a proof-of-principle examination of a brain tumor patient at 3 T. With its specificity for the mobile fraction of the proteome, its comparable sensitivity to conventional water proton MRI and its applicability to clinical MR scanners, this technique represents a further step towards the non-invasive imaging of proteomic changes in humans.


Subject(s)
Magnetic Resonance Imaging , Proteins/analysis , Humans , Macromolecular Substances/analysis , Male , Middle Aged , Signal Processing, Computer-Assisted
8.
Sci Rep ; 8(1): 1176, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29352234

ABSTRACT

Detection of multiple relaxation pools using MRI is useful in a number of neuro-pathologies including multiple sclerosis (MS), Alzheimer's, and stroke. In this study we evaluate the feasibility of using T1ρ imaging for the detection of bi-exponential decays in the human brain. A prospective T1ρ imaging study was performed on model relaxation phantoms (eggs) and 7 healthy volunteers. The data was fitted using a single pool and a 2-pool model to estimate mono- and bi-exponential T1ρ maps, respectively. Bi-exponential decays were identified in the gray matter (GM) and white matter (WM) of the brain with 40.5% of GM, and 65.1% of WM pixels showing two T1ρ relaxation pools (significance level P < 0.05). Detection of T1ρ based bi-exponential decays in the brain provides complimentary information to T2 based contrast regarding the in vivo micro-environment in the brain.


Subject(s)
Brain Mapping , Brain/pathology , Magnetic Resonance Imaging , Adult , Brain Mapping/methods , Female , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging/methods , Male , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/pathology , Phantoms, Imaging , Young Adult
9.
Radiology ; 285(3): 914-922, 2017 12.
Article in English | MEDLINE | ID: mdl-28628422

ABSTRACT

Purpose To evaluate the ability to detect intracerebral regions of increased glucose concentration at T1ρ-weighted dynamic glucose-enhanced (DGE) magnetic resonance (MR) imaging at 7.0 T. Materials and Methods This prospective study was approved by the institutional review board. Nine patients with newly diagnosed glioblastoma and four healthy volunteers were included in this study from October 2015 to July 2016. Adiabatically prepared chemical exchange-sensitive spin-lock imaging was performed with a 7.0-T whole-body unit with a temporal resolution of approximately 7 seconds, yielding the time-resolved DGE contrast. T1ρ-weighted DGE MR imaging was performed with injection of 100 mL of 20% d-glucose via the cubital vein. Glucose enhancement, given by the relative signal intensity change at T1ρ-weighted MR imaging (DGEρ), was quantitatively investigated in brain gray matter versus white matter of healthy volunteers and in tumor tissue versus normal-appearing white matter of patients with glioblastoma. The median signal intensities of the assessed brain regions were compared by using the Wilcoxon rank-sum test. Results In healthy volunteers, the median signal intensity in basal ganglia gray matter (DGEρ = 4.59%) was significantly increased compared with that in white matter tissue (DGEρ = 0.65%) (P = .028). In patients, the median signal intensity in the glucose-enhanced tumor region as displayed on T1ρ-weighted DGE images (DGEρ = 2.02%) was significantly higher than that in contralateral normal-appearing white matter (DGEρ = 0.08%) (P < .0001). Conclusion T1ρ-weighted DGE MR imaging in healthy volunteers and patients with newly diagnosed, untreated glioblastoma enabled visualization of brain glucose physiology and pathophysiologically increased glucose uptake and may have the potential to provide information about glucose metabolism in tumor tissue. © RSNA, 2017 Online supplemental material is available for this article.


Subject(s)
Brain Neoplasms/diagnostic imaging , Brain Neoplasms/metabolism , Diffusion Magnetic Resonance Imaging/methods , Glioblastoma/metabolism , Glioblastoma/pathology , Glucose/pharmacokinetics , Adult , Aged , Brain/diagnostic imaging , Brain/metabolism , Contrast Media/administration & dosage , Contrast Media/pharmacokinetics , Female , Glucose/administration & dosage , Humans , Image Enhancement , Injections, Intravenous , Male , Middle Aged , Molecular Imaging/methods , Reproducibility of Results , Sensitivity and Specificity , Young Adult
10.
Phys Rev E ; 95(2-1): 022404, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28298006

ABSTRACT

Diffusion pore imaging is an extension of diffusion-weighted nuclear magnetic resonance imaging enabling the direct measurement of the shape of arbitrarily formed, closed pores by probing diffusion restrictions using the motion of spin-bearing particles. Examples of such pores comprise cells in biological tissue or oil containing cavities in porous rocks. All pores contained in the measurement volume contribute to one reconstructed image, which reduces the problem of vanishing signal at increasing resolution present in conventional magnetic resonance imaging. It has been previously experimentally demonstrated that pore imaging using a combination of a long and a narrow magnetic field gradient pulse is feasible. In this work, an experimental verification is presented showing that pores can be imaged using short gradient pulses only. Experiments were carried out using hyperpolarized xenon gas in well-defined pores. The phase required for pore image reconstruction was retrieved from double diffusion encoded (DDE) measurements, while the magnitude could either be obtained from DDE signals or classical diffusion measurements with single encoding. The occurring image artifacts caused by restrictions of the gradient system, insufficient diffusion time, and by the phase reconstruction approach were investigated. Employing short gradient pulses only is advantageous compared to the initial long-narrow approach due to a more flexible sequence design when omitting the long gradient and due to faster convergence to the diffusion long-time limit, which may enable application to larger pores.

11.
Sci Rep ; 7: 42093, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28169369

ABSTRACT

Common medical imaging techniques usually employ contrast agents that are chemically labeled, e.g. with radioisotopes in the case of PET, iodine in the case of CT or paramagnetic metals in the case of MRI to visualize the heterogeneity of the tumor microenvironment. Recently, it was shown that natural unlabeled D-glucose can be used as a nontoxic biodegradable contrast agent in Chemical Exchange sensitive Spin-Lock (CESL) magnetic resonance imaging (MRI) to detect the glucose uptake and potentially the metabolism of tumors. As an important step to fulfill the clinical needs for practicability, reproducibility and imaging speed we present here a robust and quantitative T1ρ-weighted technique for dynamic glucose enhanced MRI (DGE-MRI) with a temporal resolution of less than 7 seconds. Applied to a brain tumor patient, the new technique provided a distinct DGE contrast between tumor and healthy brain tissue and showed the detailed dynamics of the glucose enhancement after intravenous injection. Development of this fast and quantitative DGE-MRI technique allows for a more detailed analysis of DGE correlations in the future and potentially enables non-invasive diagnosis, staging and monitoring of tumor response to therapy.


Subject(s)
Brain Neoplasms/diagnostic imaging , Brain/diagnostic imaging , Contrast Media/metabolism , Glioblastoma/diagnostic imaging , Glucose/metabolism , Image Enhancement/methods , Magnetic Resonance Imaging/methods , Aged , Brain/metabolism , Brain/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Contrast Media/pharmacokinetics , Glioblastoma/metabolism , Glioblastoma/pathology , Glucose/pharmacokinetics , Humans , Injections, Intravenous , Male , Phantoms, Imaging/statistics & numerical data , Reproducibility of Results
12.
NMR Biomed ; 30(5)2017 May.
Article in English | MEDLINE | ID: mdl-28111824

ABSTRACT

High field MRI is beneficial for chemical exchange saturation transfer (CEST) in terms of high SNR, CNR, and chemical shift dispersion. These advantages may, however, be counter-balanced by the increased transmit field inhomogeneity normally associated with high field MRI. The relatively high sensitivity of the CEST contrast to B1 inhomogeneity necessitates the development of correction methods, which is essential for the clinical translation of CEST. In this work, two B1 correction algorithms for the most studied CEST effects, amide-CEST and nuclear Overhauser enhancement (NOE), were analyzed. Both methods rely on fitting the multi-pool Bloch-McConnell equations to the densely sampled CEST spectra. In the first method, the correction is achieved by using a linear B1 correction of the calculated amide and NOE CEST effects. The second method uses the Bloch-McConnell fit parameters and the desired B1 amplitude to recalculate the CEST spectra, followed by the calculation of B1 -corrected amide and NOE CEST effects. Both algorithms were systematically studied in Bloch-McConnell equations and in human data, and compared with the earlier proposed ideal interpolation-based B1 correction method. In the low B1 regime of 0.15-0.50 µT (average power), a simple linear model was sufficient to mitigate B1 inhomogeneity effects on a par with the interpolation B1 correction, as demonstrated by a reduced correlation of the CEST contrast with B1 in both the simulations and the experiments.


Subject(s)
Amides/metabolism , Artifacts , Brain/anatomy & histology , Brain/metabolism , Magnetic Resonance Imaging/methods , Molecular Imaging/methods , Signal Processing, Computer-Assisted , Algorithms , Humans , Image Enhancement/methods , Magnetic Resonance Spectroscopy/methods , Reproducibility of Results , Sensitivity and Specificity
13.
Magn Reson Med ; 77(1): 196-208, 2017 01.
Article in English | MEDLINE | ID: mdl-26845067

ABSTRACT

PURPOSE: The chemical exchange saturation transfer (CEST) effect observed in brain tissue in vivo at the frequency offset 3.5 ppm downfield of water was assigned to amide protons of the protein backbone. Obeying a base-catalyzed exchange process such an amide-CEST effect would correlate with intracellular pH and protein concentration, correlations that are highly interesting for cancer diagnosis. However, recent experiments suggested that, besides the known aliphatic relayed-nuclear Overhauser effect (rNOE) upfield of water, an additional downfield rNOE is apparent in vivo resonating as well around +3.5 ppm. In this study, we present further evidence for the underlying downfield-rNOE signal, and we propose a first method that suppresses the downfield-rNOE contribution to the amide-CEST contrast. Thus, an isolated amide-CEST effect depending mainly on amide proton concentration and pH is generated. METHODS: The isolation of the exchange mediated amide proton effect was investigated in protein model-solutions and tissue lysates and successfully applied to in vivo CEST images of 11 glioblastoma patients. RESULTS: Comparison with gadolinium contrast enhancing longitudinal relaxation time-weighted images revealed that the downfield-rNOE-suppressed amide-CEST contrast forms a unique contrast that delineates tumor regions and show remarkable overlap with the gadolinium contrast enhancement. CONCLUSION: Thus, suppression of the downfield rNOE contribution might be the important step to yield the amide proton CEST contrast originally aimed at. Magn Reson Med 77:196-208, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Brain Neoplasms/diagnostic imaging , Glioblastoma/diagnostic imaging , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Algorithms , Brain/diagnostic imaging , Humans , Phantoms, Imaging
14.
Magn Reson Med ; 77(2): 571-580, 2017 02.
Article in English | MEDLINE | ID: mdl-26857219

ABSTRACT

PURPOSE: Together with the development of MRI contrasts that are inherently small in their magnitude, increased magnetic field accuracy is also required. Hence, mapping of the static magnetic field (B0 ) and the excitation field (B1 ) is not only important to feedback shim algorithms, but also for postprocess contrast-correction procedures. METHODS: A novel field-inhomogeneity mapping method is presented that allows simultaneous mapping of the water shift and B1 (WASABI) using an off-resonant rectangular preparation pulse. The induced Rabi oscillations lead to a sinc-like spectrum in the frequency-offset dimension and allow for determination of B0 by its symmetry axis and of B1 by its oscillation frequency. RESULTS: Stability of the WASABI method with regard to the influences of T1 , T2 , magnetization transfer, and repetition time was investigated and its convergence interval was verified. B0 and B1 maps obtained simultaneously by means of WASABI in the human brain at 3 T and 7 T can compete well with maps obtained by standard methods. Finally, the method was applied successfully for B0 and B1 correction of chemical exchange saturation transfer MRI (CEST-MRI) data of the human brain. CONCLUSION: The proposed WASABI method yields a novel simultaneous B0 and B1 mapping within 1 min that is robust and easy to implement. Magn Reson Med 77:571-580, 2017. © 2016 International Society for Magnetic Resonance in Medicine.


Subject(s)
Algorithms , Artifacts , Body Water/metabolism , Brain/metabolism , Image Interpretation, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Molecular Imaging/methods , Adult , Brain/anatomy & histology , Humans , Image Enhancement/methods , Male , Reproducibility of Results , Sensitivity and Specificity , Tissue Distribution
15.
Magn Reson Med ; 78(1): 215-225, 2017 07.
Article in English | MEDLINE | ID: mdl-27521026

ABSTRACT

PURPOSE: Chemical exchange sensitive spin-lock and related techniques allow to observe the uptake of administered D-glucose in vivo. The exchange-weighting increases with the magnetic field strength, but inhomogeneities in the radiofrequency (RF) field at ultrahigh field whole-body scanners lead to artifacts in conventional spin-lock experiments. Thus, our aim was the development of an adiabatically prepared T1ρ -based imaging sequence applicable to studies of glucose metabolism in tumor patients at ultrahigh field strengths. METHODS: An adiabatically prepared on-resonant spin-lock approach was realized at a 7 Tesla whole-body scanner and compared with conventional spin-lock. The insensitivity to RF field inhomogeneities as well as the chemical exchange sensitivity of the approach was investigated in simulations, model solutions and in the human brain. RESULTS: The suggested spin-lock approach was shown to be feasible for in vivo application at ultrahigh field whole-body scanners and showed substantially improved image quality compared with conventional spin-lock. The sensitivity of the presented method to glucose was verified in model solutions and a glucose contrast was observed in a glioblastoma patient after intravenous administration of glucose solution. CONCLUSION: An adiabatically prepared spin-lock preparation was presented that enables a homogeneous and chemical exchange sensitive T1ρ -based imaging at ultra-high field whole-body scanners, e.g., for T1ρ -based dynamic glucose enhanced MRI. Magn Reson Med 78:215-225, 2017. © 2016 International Society for Magnetic Resonance in Medicine.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Glucose/metabolism , Image Interpretation, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Molecular Imaging/methods , Algorithms , Humans , Image Enhancement/methods , Magnetic Fields , Magnetic Resonance Imaging/instrumentation , Phantoms, Imaging , Radiation Dosage , Reproducibility of Results , Sensitivity and Specificity , Spin Labels
16.
NMR Biomed ; 28(7): 906-13, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26010522

ABSTRACT

Chemical exchange saturation transfer (CEST) allows the detection of metabolites of low concentration in tissue with nearly the sensitivity of MRI with water protons. With this spectroscopic imaging approach, several tissue-specific CEST effects have been observed in vivo. Some of these originate from exchanging sites of proteins, such as backbone amide protons, or from aliphatic protons within the hydrophobic protein core. In this work, we employed CEST experiments to detect global protein unfolding. Spectral evaluation revealed exchange- and NOE-mediated CEST effects that varied in a highly characteristic manner with protein unfolding tracked by fluorescence spectroscopy. We suggest the use of this comprehensive spectral signature for the detection of protein unfolding by CEST, as it relies on several spectral hallmarks. As proof of principle, we demonstrate that the presented signature is readily detectable using a whole-body MR tomograph (B0 = 7 T), not only in denatured aqueous protein solutions, but also in heat-shocked yeast cells. A CEST imaging contrast with the potential to detect global protein unfolding would be of particular interest regarding protein unfolding as a marker for stress, ageing, and disease.


Subject(s)
Magnetic Resonance Imaging/methods , Magnetic Resonance Spectroscopy/methods , Molecular Imaging/methods , Protein Folding , Proteins/chemistry , Proteins/ultrastructure , Algorithms , Reproducibility of Results , Sensitivity and Specificity , Whole Body Imaging/methods
17.
Neuroimage ; 112: 180-188, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25727379

ABSTRACT

Endogenous chemical exchange saturation transfer (CEST) effects of protons resonating near to water protons are always diluted by competing effects such as direct water saturation and semi-solid magnetization transfer (MT). This leads to unwanted T2 and MT signal contributions that contaminate the observed CEST signal. Furthermore, all CEST effects appear to be scaled by the T1 relaxation time of the mediating water pool. As MT, T1 and T2 are also altered in tumor regions, a recently published correction algorithm yielding the apparent exchange-dependent relaxation AREX, is used to evaluate in vivo CEST effects. This study focuses on CEST effects of amides (3.5ppm) and Nuclear-Overhauser-mediated saturation transfer (NOE, -3.5ppm) that can be properly isolated at 7T. These were obtained in 10 glioblastoma patients, and this is the first comprehensive study where AREX is applied in human brain as well as in human glioblastoma. The correction of CEST effects alters the contrast significantly: after correction, the CEST effect of amides does not show significant contrast between contrast enhancing tumor regions and normal tissue, whereas NOE drops significantly in the tumor area. In addition, new features in the AREX contrasts are visible. This suggests that previous CEST approaches might not have shown pure CEST effects, but rather water relaxation shine-through effects. Our insights help to improve understanding of the CEST effect changes in tumors and correlations on a cellular and molecular level.


Subject(s)
Amides/metabolism , Brain Neoplasms/pathology , Brain/anatomy & histology , Brain/pathology , Glioblastoma/pathology , Magnetic Resonance Imaging/methods , Algorithms , Brain Chemistry , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Humans , Image Processing, Computer-Assisted , Water/metabolism
18.
PLoS One ; 10(3): e0121220, 2015.
Article in English | MEDLINE | ID: mdl-25789657

ABSTRACT

OBJECTIVE: To explore the correlation between Nuclear Overhauser Enhancement (NOE)-mediated signals and tumor cellularity in glioblastoma utilizing the apparent diffusion coefficient (ADC) and cell density from histologic specimens. NOE is one type of chemical exchange saturation transfer (CEST) that originates from mobile macromolecules such as proteins and might be associated with tumor cellularity via altered protein synthesis in proliferating cells. PATIENTS AND METHODS: For 15 patients with newly diagnosed glioblastoma, NOE-mediated CEST-contrast was acquired at 7 Tesla (asymmetric magnetization transfer ratio (MTRasym) at 3.3ppm, B1 = 0.7 µT). Contrast enhanced T1 (CE-T1), T2 and diffusion-weighted MRI (DWI) were acquired at 3 Tesla and coregistered. The T2 edema and the CE-T1 tumor were segmented. ADC and MTRasym values within both regions of interest were correlated voxelwise yielding the correlation coefficient rSpearman (rSp). In three patients who underwent stereotactic biopsy, cell density of 12 specimens per patient was correlated with corresponding MTRasym and ADC values of the biopsy site. RESULTS: Eight of 15 patients showed a weak or moderate positive correlation of MTRasym and ADC within the T2 edema (0.16≤rSp≤0.53, p<0.05). Seven correlations were statistically insignificant (p>0.05, n = 4) or yielded rSp≈0 (p<0.05, n = 3). No trend towards a correlation between MTRasym and ADC was found in CE-T1 tumor (-0.310.05, n = 6). The biopsy-analysis within CE-T1 tumor revealed a strong positive correlation between tumor cellularity and MTRasym values in two of the three patients (rSppatient3 = 0.69 and rSppatient15 = 0.87, p<0.05), while the correlation of ADC and cellularity was heterogeneous (rSppatient3 = 0.545 (p = 0.067), rSppatient4 = -0.021 (p = 0.948), rSppatient15 = -0.755 (p = 0.005)). DISCUSSION: NOE-imaging is a new contrast promising insight into pathophysiologic processes in glioblastoma regarding cell density and protein content, setting itself apart from DWI. Future studies might be based on the assumption that NOE-mediated CEST visualizes cellularity more accurately than ADC, especially in the CE-T1 tumor region.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/pathology , Diffusion Magnetic Resonance Imaging , Glioblastoma/diagnosis , Glioblastoma/pathology , Brain Neoplasms/complications , Diffusion , Edema/complications , Female , Glioblastoma/complications , Humans , Male , Middle Aged , Stereotaxic Techniques
19.
NMR Biomed ; 28(5): 529-37, 2015 May.
Article in English | MEDLINE | ID: mdl-25788155

ABSTRACT

Chemical exchange saturation transfer (CEST) imaging of endogenous agents in vivo is influenced by direct water proton saturation (spillover) and semi-solid macromolecular magnetization transfer (MT). Lorentzian fit isolation and application of the inverse metric yields the pure CEST contrast AREX, which is less affected by these processes, but still depends on the measurement technique, in particular on the irradiation amplitude B1 of the saturation pulses. This study focuses on two well-known CEST effects in the slow exchange regime originating from amide and aliphatic protons resonating at 3.5 ppm or -3.5 ppm from water protons, respectively. A B1-correction of CEST contrasts is crucial for the evaluation of data obtained in clinical studies at high field strengths with strong B1-inhomogeneities. Herein two approaches for B1-inhomogeneity correction, based on either CEST contrasts or Z-spectra, are investigated. Both rely on multiple acquisitions with different B1-values. One volunteer was examined with eight different B1-values to optimize the saturation field strength and the correction algorithm. Histogram evaluation allowed quantification of the quality of the B1-correction. Finally, the correction was applied to CEST images of a patient with oligodendroglioma WHO grade 2, and showed improvement of the image quality compared with the non-corrected CEST images, especially in the tumor region.


Subject(s)
Algorithms , Artifacts , Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Magnetic Resonance Imaging/methods , Molecular Imaging/methods , Adult , Brain Neoplasms/diagnosis , Female , Humans , Image Enhancement/methods , Male , Middle Aged , Reproducibility of Results , Sensitivity and Specificity
20.
PLoS One ; 9(8): e104181, 2014.
Article in English | MEDLINE | ID: mdl-25111650

ABSTRACT

BACKGROUND AND PURPOSE: Nuclear Overhauser Enhancement (NOE) mediated chemical exchange saturation transfer (CEST) is a novel magnetic resonance imaging (MRI) technique on the basis of saturation transfer between exchanging protons of tissue proteins and bulk water. The purpose of this study was to evaluate and compare the information provided by three dimensional NOE mediated CEST at 7 Tesla (7T) and standard MRI in glioblastoma patients. PATIENTS AND METHODS: Twelve patients with newly diagnosed histologically proven glioblastoma were enrolled in this prospective ethics committee-approved study. NOE mediated CEST contrast was acquired with a modified three-dimensional gradient-echo sequence and asymmetry analysis was conducted at 3.3 ppm (B1 = 0.7 µT) to calculate the magnetization transfer ratio asymmetry (MTR(asym)). Contrast enhanced T1 (CE-T1) and T2-weighted images were acquired at 3T and used for data co-registration and comparison. RESULTS: Mean NOE mediated CEST signal based on MTR(asym) values over all patients was significantly increased (p<0.001) in CE-T1 tumor (-1.99 ± 1.22%), tumor necrosis (-1.36 ± 1.30%) and peritumoral CEST hyperintensities (PTCH) within T2 edema margins (-3.56 ± 1.24%) compared to contralateral normal appearing white matter (-8.38 ± 1.19%). In CE-T1 tumor (p = 0.015) and tumor necrosis (p<0.001) mean MTR(asym) values were significantly higher than in PTCH. Extent of the surrounding tumor hyperintensity was smaller in eight out of 12 patients on CEST than on T2-weighted images, while four displayed at equal size. In all patients, isolated high intensity regions (0.40 ± 2.21%) displayed on CEST within the CE-T1 tumor that were not discernible on CE-T1 or T2-weighted images. CONCLUSION: NOE mediated CEST Imaging at 7 T provides additional information on the structure of peritumoral hyperintensities in glioblastoma and displays isolated high intensity regions within the CE-T1 tumor that cannot be acquired on CE-T1 or T2-weighted images. Further research is needed to determine the origin of NOE mediated CEST and possible clinical applications such as therapy assessment or biopsy planning.


Subject(s)
Glioblastoma/diagnosis , Contrast Media , Female , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging/methods , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...