1
|
Mohanta Z, Stabinska J, Gilad AA, Barker PB, McMahon MT. The proton resonance enhancement for CEST imaging and shift exchange (PRECISE) family of RF pulse shapes for CEST MRI. Magn Reson Med 2025; 93:1954-1968. [PMID: 39831452 PMCID: PMC11893253 DOI: 10.1002/mrm.30410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025]
Abstract
PURPOSE To optimize a 100 ms pulse for producing CEST MRI contrast and evaluate in mice. METHODS A gradient ascent algorithm was employed to generate a family of 100 point, 100 ms pulses for use in CEST pulse trains (proton resonance enhancement for CEST imaging and shift exchange). Gradient ascent optimizations were performed for exchange rates = 500, 1500, 2500, 3500, and 4500 s-1; and labile proton offsets (Δω) = 9.6, 7.8, 4.2, and 2.0 ppm. Seven proton resonance enhancement for CEST imaging and shift exchange pulse shapes were tested on an 11.7 T scanner using a phantom containing three representative CEST agents with peak saturation B1,peak = 4 μT. The pulse producing the most contrast in phantoms was then evaluated for CEST MRI pH mapping of the kidneys in healthy mice after iopamidol administration. RESULTS The most promising pulse in terms of contrast performance across all three phantoms was the 9.6 ppm, 2500 s-1 optimized pulse with ˜2.7 × increase in asymmetric magnetization transfer ratio (MTRasym) over Gaussian, and ˜ 1.3 times over Fermi pulses for the same B1,peak = 4 μT. This pulse also displayed a large improvement in contrast over the Gaussian pulse after administration of iopamidol in live mice. CONCLUSION A new 100-ms pulse was developed based on gradient ascent optimizations, which produced better contrast compared to standard Gaussian and Fermi pulses in phantoms. This shape also showed a substantial improvement for CEST MRI pH mapping in live mice over the Gaussian shape and appears promising for a wide range of CEST applications.
Collapse
Affiliation(s)
- Zinia Mohanta
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
| | - Julia Stabinska
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
| | - Assaf A. Gilad
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI
- Department of Radiology, Michigan State University, East Lansing, MI
| | - Peter B. Barker
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
| | - Michael T. McMahon
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
| |
Collapse
|
2
|
Singh M, Kang B, Mahmud SZ, van Zijl P, Zhou J, Heo HY. Saturation transfer MR fingerprinting for magnetization transfer contrast and chemical exchange saturation transfer quantification. Magn Reson Med 2025. [PMID: 40228056 DOI: 10.1002/mrm.30532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/16/2025]
Abstract
PURPOSE The aim of this study was to develop a saturation transfer MR fingerprinting (ST-MRF) technique using a biophysics model-driven deep learning approach. METHODS A deep learning-based quantitative saturation transfer framework was proposed to estimate water, magnetization transfer contrast, and amide proton transfer (APT) parameters plus B0 field inhomogeneity. This framework incorporated a Bloch-McConnell simulator during neural network training and enforced consistency between synthesized MRF signals and experimentally acquired ST-MRF signals. Ground-truth numerical phantoms were used to assess the accuracy of estimated tissue parameters, and in vivo tissue parameters were validated using synthetic MRI analysis. RESULTS The proposed ST-MRF reconstruction network achieved a normalized root mean square error (nRMSE) of 9.3% when tested against numerical phantoms with a signal-to-noise ratio of 46 dB, which outperformed conventional Bloch-McConnell fitting (nRMSE of 15.3%) and dictionary-matching approaches (nRMSE of 19.5%). Synthetic MRI analysis indicated excellent similarity (RMSE = 3.2%) between acquired and synthesized ST-MRF images, demonstrating high in vivo reconstruction accuracy. In healthy human brains, the APT pool size ratios for gray and white matter were 0.16 ± 0.02% and 0.13 ± 0.02%, respectively, and the exchange rates for gray and white matter were 101 ± 25 Hz and 131 ± 27 Hz, respectively. The reconstruction network processed the eight tissue parameter maps in approximately 27 s for ST-MRF data sized at 256 × 256 × 9 × 103. CONCLUSION This study highlights the feasibility of the deep learning-based ST-MRF imaging for rapid and accurate quantification of free bulk water, magnetization transfer contrast, APT parameters, and B0 field inhomogeneity.
Collapse
Affiliation(s)
- Munendra Singh
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Beomgu Kang
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sultan Z Mahmud
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Peter van Zijl
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Jinyuan Zhou
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hye-Young Heo
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Mulugeta E, Tegafaw T, Liu Y, Zhao D, Baek A, Kim J, Chang Y, Lee GH. Synthesis, Characterization, Magnetic Properties, and Applications of Carbon Dots as Diamagnetic Chemical Exchange Saturation Transfer Magnetic Resonance Imaging Contrast Agents: A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:542. [PMID: 40214587 PMCID: PMC11990683 DOI: 10.3390/nano15070542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025]
Abstract
Carbon dots (CDs) are metal-free carbon-based nanoparticles. They possess excellent photoluminescent properties, various physical properties, good chemical stability, high water solubility, high biocompatibility, and tunable surface functionalities, suitable for biomedical applications. Their properties are subject to synthetic conditions such as pH, reaction time, temperature, precursor, and solvent. Until now, a large number of articles on the synthesis and biomedical applications of CDs using their photoluminescent properties have been reported. However, their research on magnetic properties and especially, diamagnetic chemical exchange saturation transfer (diaCEST) in magnetic resonance imaging (MRI) is very poor. The diaCEST MRI contrast agents are based on exchangeable protons of materials with bulk water protons and thus, different from conventional MRI contrast agents, which are based on enhancements of proton spin relaxations of bulk water and tissue. In this review, various syntheses, characterizations, magnetic properties, and potential applications of CDs as diaCEST MRI contrast agents are reviewed. Finally, future perspectives of CDs as the next-generation diaCEST MRI contrast agents are discussed.
Collapse
Affiliation(s)
- Endale Mulugeta
- Department of Chemistry, College of Natural Sciences, Kyungpook National University, Taegu 41566, Republic of Korea; (E.M.); (T.T.); (Y.L.); (D.Z.)
| | - Tirusew Tegafaw
- Department of Chemistry, College of Natural Sciences, Kyungpook National University, Taegu 41566, Republic of Korea; (E.M.); (T.T.); (Y.L.); (D.Z.)
| | - Ying Liu
- Department of Chemistry, College of Natural Sciences, Kyungpook National University, Taegu 41566, Republic of Korea; (E.M.); (T.T.); (Y.L.); (D.Z.)
| | - Dejun Zhao
- Department of Chemistry, College of Natural Sciences, Kyungpook National University, Taegu 41566, Republic of Korea; (E.M.); (T.T.); (Y.L.); (D.Z.)
| | - Ahrum Baek
- Institute of Biomedical Engineering Research, Kyungpook National University, Taegu 41944, Republic of Korea;
| | - Jihyun Kim
- Department of Chemistry Education, Teachers’ College, Kyungpook National University, Taegu 41566, Republic of Korea;
| | - Yongmin Chang
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Taegu 41944, Republic of Korea
| | - Gang Ho Lee
- Department of Chemistry, College of Natural Sciences, Kyungpook National University, Taegu 41566, Republic of Korea; (E.M.); (T.T.); (Y.L.); (D.Z.)
| |
Collapse
|
4
|
Vladimirov N, Cohen O, Heo HY, Zaiss M, Farrar CT, Perlman O. Quantitative molecular imaging using deep magnetic resonance fingerprinting. Nat Protoc 2025:10.1038/s41596-025-01152-w. [PMID: 40169753 DOI: 10.1038/s41596-025-01152-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 01/17/2025] [Indexed: 04/03/2025]
Abstract
Deep learning-based saturation transfer magnetic resonance fingerprinting (MRF) is an emerging approach for noninvasive in vivo imaging of proteins, metabolites and pH. It involves a series of steps, including sample/participant preparation, image acquisition schedule design, biophysical model formulation and artificial intelligence and computational model training, followed by image acquisition, deep reconstruction and analysis. Saturation transfer-based molecular MRI has been slow to reach clinical maturity and adoption for clinical practice due to its technical complexity, semi-quantitative contrast-weighted nature and long scan times needed for the extraction of quantitative molecular biomarkers. Deep MRF provides solutions to these challenges by providing a quantitative and rapid framework for extracting biologically and clinically meaningful molecular information. Here we define a complete protocol for quantitative molecular MRI using deep MRF. We describe in vitro sample preparation and animal and human scan considerations, and provide intuition behind the acquisition protocol design and optimization of chemical exchange saturation transfer (CEST) and semi-solid magnetization transfer (MT) quantitative imaging. We then extensively describe the building blocks for several artificial intelligence models and demonstrate their performance for different applications, including cancer monitoring, brain myelin imaging and pH quantification. Finally, we provide guidelines to further modify and expand the pipeline for imaging a variety of other pathologies (such as neurodegeneration, stroke and cardiac disease), accompanied by the related open-source code and sample data. The procedure takes between 48 min (for two proton pools or in vitro imaging) and 57 h (for complex multi-proton pool in vivo imaging) to complete and is suitable for graduate student-level users.
Collapse
Affiliation(s)
- Nikita Vladimirov
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Ouri Cohen
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hye-Young Heo
- Divison of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Moritz Zaiss
- Institute of Neuroradiology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
- Department of Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian T Farrar
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Or Perlman
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel.
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
5
|
Zu T, Yong X, Dai Z, Jiang T, Hsu YC, Lu S, Zhang Y. Prospective acceleration of whole-brain CEST imaging by golden-angle view ordering in Cartesian coordinates and joint k-space and image-space parallel imaging (KIPI). Magn Reson Med 2025; 93:1585-1601. [PMID: 39607875 DOI: 10.1002/mrm.30375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/16/2024] [Accepted: 10/29/2024] [Indexed: 11/30/2024]
Abstract
PURPOSE To prospectively accelerate whole-brain CEST acquisition by joint k-space and image-space parallel imaging (KIPI) with a proposed golden-angle view ordering technique (GAVOT) in Cartesian coordinates. THEORY AND METHODS The T2-decay effect will vary across frames with variable acceleration factors (AF) in the prospective acquisition using sequences with long echo trains. The GAVOT method uses a subset strategy to eliminate the T2-decay inconsistency, where all frames use a subset of shots from the calibration frame to form their k-space view ordering. The golden-angle rule is adapted to ensure uniform k-space coverage for arbitrary AFs in Cartesian coordinates. Phantom and in vivo studies were conducted on a 3 T scanner. RESULTS The GAVOT view ordering yielded a higher g-factor than conventional uniformly centric ordering, whereas the noise propagation in amide proton transfer (APT) weighted images was similar between different view ordering. Compared to centric ordering, GAVOT successfully eliminated the T2-decay inconsistency across all frames, resulting in fewer image artifacts for both KIPI and conventional parallel imaging methods. The synergy of GAVOT and KIPI mitigated strong aliasing artifacts and achieved high-quality reconstruction of prospective variable-AF datasets. GAVOT-KIPI reduced the scan time to 2.1 min for whole-brain APT weighted imaging and 4.7 min for quantitative APT signal (APT#) mapping. CONCLUSION GAVOT makes the prospective variable AF strategy flexible and practical, and, in conjunction with KIPI, ensures high-quality reconstruction from highly undersampled data, facilitating the clinical translation of whole-brain CEST imaging.
Collapse
Affiliation(s)
- Tao Zu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Xingwang Yong
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Zhechuan Dai
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Tongling Jiang
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Yi-Cheng Hsu
- MR Collaboration, Siemens Healthcare, Shanghai, China
| | - Shanshan Lu
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi Zhang
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
6
|
Wang Q, Zhao C, Wang S, Wang J, Wu F, Ombrini P, Ganapathy S, Eustace S, Bai X, Li B, Armand M, Aurbach D, Wagemaker M. Interphase Design for Lithium-Metal Anodes. J Am Chem Soc 2025; 147:9365-9377. [PMID: 40053684 DOI: 10.1021/jacs.4c15759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2025]
Abstract
Electrode-electrolyte interphases are critical determinants of the reversibility and longevity of lithium (Li)-metal batteries (LMBs). However, upon cycling, the inherently delicate interphases, formed from electrolyte decomposition, become vulnerable to chemomechanical degradation and corrosion, resulting in rapid capacity loss and thus short battery life. Here, we present a comprehensive analysis of the complex interplay between the thermodynamic and kinetic properties of interphases on Li-metal anodes, providing insights into interphase design to address these challenges. Direct measurements of ion-transport kinetics across various electrolyte chemistries reveal that interphases with high Li-ion mobility are essential for achieving dense Li deposits. Conversely, sluggish ion transport generates high-surface-area Li deposits that induce Li random stripping and the accumulation of isolated Li deposits. Surprisingly, interphases that support long cycle life do not necessarily require the formation of dense Li deposits but must avoid possible electrochemical/chemical reactions between the Li-metal deposits and electrolytes' components. By that, in some specific electrolyte systems, isolated Li deposits can recover and electrically rejoin the active Li anodes' mass. These findings challenge conventional understanding and establish new principles for designing durable LMBs, demonstrating that even with commercial carbonate-based electrolytes, LiNi0.8Co0.1Mn0.1O2||Cu cells can achieve high reversibility.
Collapse
Affiliation(s)
- Qidi Wang
- Department of Radiation Science and Technology, Delft University of Technology, Delft 2629JB, The Netherlands
| | - Chenglong Zhao
- Department of Radiation Science and Technology, Delft University of Technology, Delft 2629JB, The Netherlands
| | - Shuwei Wang
- Shenzhen Key Laboratory on Power Battery Safety and Shenzhen Geim Graphene Center, School of Shenzhen International Graduate, Tsinghua University, Beijing, Guangdong 518055, China
| | - Jianlin Wang
- State Key Laboratory for Surface Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
| | - Fangting Wu
- Shenzhen Key Laboratory on Power Battery Safety and Shenzhen Geim Graphene Center, School of Shenzhen International Graduate, Tsinghua University, Beijing, Guangdong 518055, China
| | - Pierfrancesco Ombrini
- Department of Radiation Science and Technology, Delft University of Technology, Delft 2629JB, The Netherlands
| | - Swapna Ganapathy
- Department of Radiation Science and Technology, Delft University of Technology, Delft 2629JB, The Netherlands
| | - Stephen Eustace
- Department of Biotechnology, Delft University of Technology, Delft 2629HZ, The Netherlands
| | - Xuedong Bai
- State Key Laboratory for Surface Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
| | - Baohua Li
- Shenzhen Key Laboratory on Power Battery Safety and Shenzhen Geim Graphene Center, School of Shenzhen International Graduate, Tsinghua University, Beijing, Guangdong 518055, China
| | - Michel Armand
- Centre for Cooperative Research on Alternative Energies (CIC energiGUNE), Basque Research and Technology Alliance (BRTA), 01510 Vitoria-Gasteiz, Spain
| | - Doron Aurbach
- Chemistry Department, BINA-BIU Center for Nanotechnology and Advanced Materials and INIES-Israel National Institute for Energy Storage, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Marnix Wagemaker
- Department of Radiation Science and Technology, Delft University of Technology, Delft 2629JB, The Netherlands
| |
Collapse
|
7
|
Scarciglia A, Papi C, Romiti C, Leone A, Di Gregorio E, Ferrauto G. Gadolinium-Based Contrast Agents (GBCAs) for MRI: A Benefit-Risk Balance Analysis from a Chemical, Biomedical, and Environmental Point of View. GLOBAL CHALLENGES (HOBOKEN, NJ) 2025; 9:2400269. [PMID: 40071223 PMCID: PMC11891575 DOI: 10.1002/gch2.202400269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 01/07/2025] [Indexed: 03/14/2025]
Abstract
Gadolinium-based contrast agents (GBCAs) have revolutionized medical imaging, enhancing the accuracy and diagnostic value of magnetic resonance imaging (MRI). The increasing use of GBCAs has raised concerns about the release of gadolinium (Gd)(III) into the environment and potential risks for human health. Initially, multiple administrations of GBCAs were associated only with nephrogenic system fibrosis disease in individuals with impaired kidney function. Even if the Gd(III) retention in tissues has not yet been correlated with any specific disease, caution is required for the extensive use of GBCAs. The concerns related to the employment of GBCAs, due to the possible deposition and retention, should be extended also to healthy individuals without renal impairments. To ensure the well-being of patients, there is a need to develop even more stable and better-performing GBCAs, new MRI approaches requiring lower doses of GBCAs and, finally, innovative methods for recovering Gd(III) from both patients' urines and the environment. This can have strong advantages for human health and for environmental sustainability, also considering Gd(III) scarcity, being a rare earth element, and the shared guideline to reduce, as much as possible, the use of rare metals.
Collapse
Affiliation(s)
- Angelo Scarciglia
- Department of Molecular Biotechnologies and Health SciencesUniversity of TorinoVia Nizza 52Torino10126Italy
| | - Chiara Papi
- Department of Molecular Biotechnologies and Health SciencesUniversity of TorinoVia Nizza 52Torino10126Italy
| | - Chiara Romiti
- Department of Molecular Biotechnologies and Health SciencesUniversity of TorinoVia Nizza 52Torino10126Italy
| | - Andrea Leone
- Department of Molecular Biotechnologies and Health SciencesUniversity of TorinoVia Nizza 52Torino10126Italy
| | - Enza Di Gregorio
- Department of Molecular Biotechnologies and Health SciencesUniversity of TorinoVia Nizza 52Torino10126Italy
| | - Giuseppe Ferrauto
- Department of Molecular Biotechnologies and Health SciencesUniversity of TorinoVia Nizza 52Torino10126Italy
| |
Collapse
|
8
|
Yang H, Wu Q, Li L, Wu Y. Evaluation of Brain Impairment Using Proton Exchange Rate MRI in a Kainic Acid-Induced Rat Model of Epilepsy. Mol Imaging Biol 2025; 27:1-9. [PMID: 39747781 DOI: 10.1007/s11307-024-01980-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 11/27/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025]
Abstract
PURPOSE Proton exchange rate (Kex) is a valuable biophysical metric. Kex MRI may augment conventional structural MRI by revealing brain impairments at the molecular level. This study aimed to investigate the feasibility of Kex MRI in evaluating brain injuries at multiple epilepsy stages. PROCEDURES Six adult rats with epilepsy induced by intra-amygdalae administration of kainic acid (KA) underwent MRI experiment at 11.7 T. Two MRI scans, including T1 mapping and CEST imaging under three B1 amplitudes of 0.75, 1.0, and 1.5 μT, were conducted before and 2, 7, and 28 days after KA injection. Quasi-steady-state analysis was performed to reconstruct equilibrium Z spectra. Direct saturation was resolved using a multi-pool Lorentzian model and removed from Z spectra. The residual spectral signal (ΔZ) was used to construct the omega plot of (1-ΔZ)/ΔZ as a linear function of 1/ ω 1 2 , from which Kex was quantified from the X-axis intercept. One-way ANOVA or two-tailed paired student's t-test was employed with P < 0.05 as statistically significant. RESULTS All animals exhibited repetitive status epilepticus with IV to V seizure stages after KA injection. At day 28, Kex values in the hippocampus and cerebral cortex at the surgical hemisphere with KA injection were significantly higher than that at the time points of control and/or day 2 in the same regions (P < 0.01). Moreover, the values were significantly higher than that in respective contralateral regions at day 28 (P < 0.02). No substantial changes of Kex were seen in bilateral thalamus or contralateral hemisphere among time points (all P > 0.05). CONCLUSIONS Kex increase significantly in the cerebral cortex and hippocampus at the surgical hemisphere, especially at day 28, likely due to substantial alterations at chronic epilepsy stage. Kex MRI is promising to evaluate brain impairment, facilitating the diagnosis and evaluation of neurological disorders.
Collapse
Affiliation(s)
- Huanhuan Yang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Qiting Wu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Lin Li
- Surgery Division, Epilepsy Center, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Yin Wu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China.
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, Guangdong, China.
| |
Collapse
|
9
|
Lee RC, Boparai MS, Duong TQ. Detection of breast cancer lesions using APT weighted MRI: a systematic review. J Transl Med 2025; 23:141. [PMID: 39891260 PMCID: PMC11786454 DOI: 10.1186/s12967-025-06153-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/18/2025] [Indexed: 02/03/2025] Open
Abstract
Amide proton transfer (APT) is a novel magnetic resonance imaging (MRI) technique that has shown promising ability to study cancers. This paper systematically reviewed the literature on the use of APT MRI in the prognosis of breast cancer. A literature search was conducted on Pubmed and Embase and a total of 14 articles comprising 775 patients were included in the review. APT MRI had the ability to distinguish between benign and malignant lesions with an AUC as high as 0.959. There is a positive correlation between APT signal intensity and tumor grade/stage as well as Ki-67, whereas no correlation was found with ER/PR/Her-2 receptor status. There was a greater decrease in APT signal intensity after neoadjuvant chemotherapy (NAC) in responders compared to non-responders, suggesting that APT MRI may serve as a valuable supplemental tool in the early identification of chemotherapy response. APT has the potential to complement with other imaging methods in the diagnosis, prognosis, treatment monitoring, and management of breast cancer. Additional studies and standardization of APT acquisition methods are needed.
Collapse
Affiliation(s)
- Ryan C Lee
- Department of Radiology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA
| | - Montek Singh Boparai
- Department of Radiology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA
| | - Tim Q Duong
- Department of Radiology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
10
|
Sun D, Wu G, Zhang W, Gharaibeh NM, Li X. Visualizing Preosteoarthritis: Updates on UTE-Based Compositional MRI and Deep Learning Algorithms. J Magn Reson Imaging 2025. [PMID: 39792443 DOI: 10.1002/jmri.29710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025] Open
Abstract
Osteoarthritis (OA) is heterogeneous and involves structural changes in the whole joint, such as cartilage, meniscus/labrum, ligaments, and tendons, mainly with short T2 relaxation times. Detecting OA before the onset of irreversible changes is crucial for early proactive management and limit growing disease burden. The more recent advanced quantitative imaging techniques and deep learning (DL) algorithms in musculoskeletal imaging have shown great potential for visualizing "pre-OA." In this review, we first focus on ultrashort echo time-based magnetic resonance imaging (MRI) techniques for direct visualization as well as quantitative morphological and compositional assessment of both short- and long-T2 musculoskeletal tissues, and second explore how DL revolutionize the way of MRI analysis (eg, automatic tissue segmentation and extraction of quantitative image biomarkers) and the classification, prediction, and management of OA. PLAIN LANGUAGE SUMMARY: Detecting osteoarthritis (OA) before the onset of irreversible changes is crucial for early proactive management. OA is heterogeneous and involves structural changes in the whole joint, such as cartilage, meniscus/labrum, ligaments, and tendons, mainly with short T2 relaxation times. Ultrashort echo time-based magnetic resonance imaging (MRI), in particular, enables direct visualization and quantitative compositional assessment of short-T2 tissues. Deep learning is revolutionizing the way of MRI analysis (eg, automatic tissue segmentation and extraction of quantitative image biomarkers) and the detection, classification, and prediction of disease. They together have made further advances toward identification of imaging biomarkers/features for pre-OA. LEVEL OF EVIDENCE: 5 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Dong Sun
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Wu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Zhang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nadeer M Gharaibeh
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoming Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Sun PZ. Physics-guided multi-dimensional scan optimization and quasi-steady-state reconstruction to enhance CEST MRI sensitivity efficiency and quantification accuracy. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2025; 370:107821. [PMID: 39689390 PMCID: PMC11725439 DOI: 10.1016/j.jmr.2024.107821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 11/20/2024] [Accepted: 12/07/2024] [Indexed: 12/19/2024]
Abstract
Chemical exchange saturation transfer (CEST) MRI has become increasingly utilized for detecting dilute labile protons and characterizing microenvironment properties. However, the CEST MRI effect is only a few percent, and there is a need for a systematic approach to optimize scan parameters for sensitive and accurate CEST quantification. We propose multi-dimensional adjustments of key parameters such as the repetition time (TR) and RF duty cycle to optimize CEST MRI sensitivity per unit of time and utilization of quasi-steady-state (QUASS) reconstruction to recover the full CEST effect during postprocessing. Our work herein derived the CEST effect based on the generalized spin-lock CEST model and determined the interdependency of the optimal RF duty cycle and TR, showing the optimal TR decreases with the RF duty cycle but plateaus beyond 60-80 %. The accuracy of the solution was validated with both numerical simulations and CEST MRI experiments on a dual pH creatine gel phantom. The desired equilibrium CEST effect was further reconstructed with the QUASS algorithm from the optimized CEST MRI scan. In summary, our study establishes a workflow for CEST MRI scan optimization and postprocessing analysis, providing a framework to boost both the sensitivity of CEST MRI scans and the accuracy of CEST quantification. This approach holds promise for future in vivo validation and translation.
Collapse
Affiliation(s)
- Phillip Zhe Sun
- Non-Human-Primate Imaging Center, Emory National Primate Research Center, Emory University, Atlanta, GA, United States; Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States; Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, United States.
| |
Collapse
|
12
|
Gujral J, Gandhi OH, Singh SB, Ahmed M, Ayubcha C, Werner TJ, Revheim ME, Alavi A. PET, SPECT, and MRI imaging for evaluation of Parkinson's disease. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2024; 14:371-390. [PMID: 39840378 PMCID: PMC11744359 DOI: 10.62347/aicm8774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 12/09/2024] [Indexed: 01/23/2025]
Abstract
This review assesses the primary neuroimaging techniques used to evaluate Parkinson's disease (PD) - a neurological condition characterized by gradual dopamine-producing nerve cell degeneration. The neuroimaging techniques explored include positron emission tomography (PET), single-photon emission computed tomography (SPECT), and magnetic resonance imaging (MRI). These modalities offer varying degrees of insights into PD pathophysiology, diagnostic accuracy, specificity by way of exclusion of other Parkinsonian syndromes, and monitoring of disease progression. Neuroimaging is thus crucial for diagnosing and managing PD, with integrated multimodal approaches and novel techniques further enhancing early detection and treatment evaluation.
Collapse
Affiliation(s)
- Jaskeerat Gujral
- Department of Radiology, University of PennsylvaniaPhiladelphia, PA 19104, USA
| | - Om H Gandhi
- Department of Radiology, University of PennsylvaniaPhiladelphia, PA 19104, USA
| | - Shashi B Singh
- Stanford University School of MedicineStanford, CA 94305, USA
| | - Malia Ahmed
- Department of Radiology, University of PennsylvaniaPhiladelphia, PA 19104, USA
| | - Cyrus Ayubcha
- Harvard Medical SchoolBoston, MA 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public HealthBoston, MA 02115, USA
| | - Thomas J Werner
- Department of Radiology, University of PennsylvaniaPhiladelphia, PA 19104, USA
| | - Mona-Elisabeth Revheim
- The Intervention Center, Rikshopitalet, Division of Technology and Innovation, Oslo University HospitalOslo 0372, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of OsloOslo 0315, Norway
| | - Abass Alavi
- Department of Radiology, University of PennsylvaniaPhiladelphia, PA 19104, USA
| |
Collapse
|
13
|
Zhu D, Fu X, Liu J, Liu X, Cheng L, Zhang X, Lu J, Qin Q, Sun P, Zhou Z, Feng Y, Wang J. Multiparametric Chemical Exchange Saturation Transfer MRI Detects Metabolic Changes in Mild Cognitive Impairment Cases at 3.0 Tesla. Neurochem Res 2024; 50:51. [PMID: 39648256 DOI: 10.1007/s11064-024-04307-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/08/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024]
Abstract
This study aimed to assess the potential of multiparametric chemical exchange saturation transfer magnetic resonance imaging (CEST MRI) for MCI detection. Twenty-eight patients with MCI and 31 age- and gender-matched normal controls (NCs) were enrolled. CEST MRI was performed with a gradient and spin-echo sequence on a 3T scanner. Multi-parametric CEST parameters were analyzed, such as amide CEST, amine CEST, amine and amide concentration independent assay (AACID), magnetization transfer ratio yielding rex (MTRrex-amide), and downfield rNOE suppressed apparent exchange-dependent relaxation amide proton (DNS-AREX-amide). Statistical analyses of CEST parameters were performed to evaluate group differences, their correlations with Montreal cognitive assessment (MoCA) score, and diagnostic performance for MCI. Compared with NC group, amide CEST as well as MTRrex-amide decreased in the left hippocampus and amine CEST as well as AACID increased in the right hippocampus in the MCI group; In both hippocampi, the DNS-AREX-amide were significantly lower in the MCI group versus the NC group (all P < 0.05). Amine CEST in the right hippocampus was negatively correlated with MoCA score (r = - 0.457, p = 0.017); DNS-AREX-amide in the bilateral hippocampus was positively correlated with MoCA score (left: r = 0.449, P = 0.019; right: AUC = 0.529, P = 0.05). DNS-AREX-amide in the bilateral hippocampus have a good ability to identify MCI (left: AUC = 0.756, P < 0.01; right: AUC = 0.762, P < 0.01). CEST MRI provides a potential imaging diagnostic strategy for MCI, which may promote early detection of MCI and provide novel insights into the pathological progress toward AD.
Collapse
Affiliation(s)
- Dongyong Zhu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xiaona Fu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Jia Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xiaoming Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Lan Cheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xinli Zhang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Jue Lu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Qian Qin
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Peng Sun
- Clinical & Technical Support, Philips Healthcare, Beijing, China
| | - Zhenyu Zhou
- Clinical & Technical Support, Philips Healthcare, Beijing, China
| | - Yiming Feng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Jing Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| |
Collapse
|
14
|
Yan M, Bie C, Jia W, Liu C, He X, Song X. Synthesis of higher-B 0 CEST Z-spectra from lower-B 0 data via deep learning and singular value decomposition. NMR IN BIOMEDICINE 2024; 37:e5221. [PMID: 39113170 DOI: 10.1002/nbm.5221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 11/15/2024]
Abstract
Chemical exchange saturation transfer (CEST) MRI at 3 T suffers from low specificity due to overlapping CEST effects from multiple metabolites, while higher field strengths (B0) allow for better separation of Z-spectral "peaks," aiding signal interpretation and quantification. However, data acquisition at higher B0 is restricted by equipment access, field inhomogeneity and safety issues. Herein, we aim to synthesize higher-B0 Z-spectra from readily available data acquired with 3 T clinical scanners using a deep learning framework. Trained with simulation data using models based on Bloch-McConnell equations, this framework comprised two deep neural networks (DNNs) and a singular value decomposition (SVD) module. The first DNN identified B0 shifts in Z-spectra and aligned them to correct frequencies. After B0 correction, the lower-B0 Z-spectra were streamlined to the second DNN, casting into the key feature representations of higher-B0 Z-spectra, obtained through SVD truncation. Finally, the complete higher-B0 Z-spectra were recovered from inverse SVD, given the low-rank property of Z-spectra. This study constructed and validated two models, a phosphocreatine (PCr) model and a pseudo-in-vivo one. Each experimental dataset, including PCr phantoms, egg white phantoms, and in vivo rat brains, was sequentially acquired on a 3 T human and a 9.4 T animal scanner. Results demonstrated that the synthetic 9.4 T Z-spectra were almost identical to the experimental ground truth, showing low RMSE (0.11% ± 0.0013% for seven PCr tubes, 1.8% ± 0.2% for three egg white tubes, and 0.79% ± 0.54% for three rat slices) and high R2 (>0.99). The synthesized amide and NOE contrast maps, calculated using the Lorentzian difference, were also well matched with the experiments. Additionally, the synthesis model exhibited robustness to B0 inhomogeneities, noise, and other acquisition imperfections. In conclusion, the proposed framework enables synthesis of higher-B0 Z-spectra from lower-B0 ones, which may facilitate CEST MRI quantification and applications.
Collapse
Affiliation(s)
- Mengdi Yan
- School of Information Sciences and Technology, Northwest University, Xi'an, China
- Center for Biomedical Imaging Research, Tsinghua University, Beijing, China
| | - Chongxue Bie
- School of Information Sciences and Technology, Northwest University, Xi'an, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wentao Jia
- School of Information Sciences and Technology, Northwest University, Xi'an, China
| | - Chuyu Liu
- Center for Biomedical Imaging Research, Tsinghua University, Beijing, China
| | - Xiaowei He
- School of Information Sciences and Technology, Northwest University, Xi'an, China
| | - Xiaolei Song
- Center for Biomedical Imaging Research, Tsinghua University, Beijing, China
| |
Collapse
|
15
|
Wang Q, Zhao C, Hu X, Wang J, Ganapathy S, Eustace S, Bai X, Li B, Li H, Aurbach D, Wagemaker M. Grain-Boundary-Rich Interphases for Rechargeable Batteries. J Am Chem Soc 2024; 146:31778-31787. [PMID: 39513539 PMCID: PMC11583303 DOI: 10.1021/jacs.4c10650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/15/2024]
Abstract
The formation of stable interphases on the electrodes is crucial for rechargeable lithium (Li) batteries. However, next-generation high-energy batteries face challenges in controlling interphase formation due to the high reactivity and structural changes of electrodes, leading to reduced stability and slow ion transport, which accelerate battery degradation. Here, we report an approach to address these issues by introducing multicomponent grain-boundary-rich interphase that boosts the rapid transport of ions and enhances passivation toward prolonged lifespan. This is guided by fundamental principles of solid-state ionics and geological crystallization differentiation theory, achieved through improved solvation chemistry. Demonstrations showcase how the introduction of the interphase substantially impacts the Li-ion transport across the interphase and the electrode-electrolyte compatibility in cost-effective electrolyte solutions optimized with multiple Li salts. The resulting interphases feature microstructures rich in inorganic grain boundaries with a diverse array of nanosized grains, presenting enhanced Li-ion transport. Comprehensive analyses revealed that this realizes remarkable electrochemical stability over extended cycling periods by inhibiting electrode corrosion, thus holding promise for high-capacity thin-Li-metal, Si-based anodes, and even Li-free anodes when paired with high-capacity oxide cathodes. This work opens new avenues to customize protective interphases on high-capacity electrodes, promoting the development of batteries with the highest energy density using cost-effective electrolytes.
Collapse
Affiliation(s)
- Qidi Wang
- Department
of Radiation Science and Technology, Delft
University of Technology, Delft 2629 JB, The Netherlands
| | - Chenglong Zhao
- Department
of Radiation Science and Technology, Delft
University of Technology, Delft 2629 JB, The Netherlands
| | - Xia Hu
- Shenzhen
Key Laboratory on Power Battery Safety and Shenzhen Geim Graphene
Center, Shenzhen International Graduate School, Tsinghua University, Guangdong 518055, China
| | - Jianlin Wang
- State
Key Laboratory for Surface Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
| | - Swapna Ganapathy
- Department
of Radiation Science and Technology, Delft
University of Technology, Delft 2629 JB, The Netherlands
| | - Stephen Eustace
- Department
of Biotechnology, Delft University of Technology, Delft 2629 HZ, The Netherlands
| | - Xuedong Bai
- State
Key Laboratory for Surface Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
| | - Baohua Li
- Shenzhen
Key Laboratory on Power Battery Safety and Shenzhen Geim Graphene
Center, Shenzhen International Graduate School, Tsinghua University, Guangdong 518055, China
| | - Hong Li
- Key
Laboratory for Renewable Energy, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
| | - Doron Aurbach
- Chemistry
Department, BINA-BIU Center for Nanotechnology & Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Marnix Wagemaker
- Department
of Radiation Science and Technology, Delft
University of Technology, Delft 2629 JB, The Netherlands
| |
Collapse
|
16
|
Power I, Rivlin M, Shmuely H, Zaiss M, Navon G, Perlman O. In vivo mapping of the chemical exchange relayed nuclear Overhauser effect using deep magnetic resonance fingerprinting. iScience 2024; 27:111209. [PMID: 39569380 PMCID: PMC11576397 DOI: 10.1016/j.isci.2024.111209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 09/17/2024] [Accepted: 10/16/2024] [Indexed: 11/22/2024] Open
Abstract
Noninvasive magnetic resonance imaging (MRI) of the relayed nuclear Overhauser effect (rNOE) constitutes a promising approach for gaining biological insights into various pathologies, including brain cancer, kidney injury, ischemic stroke, and liver disease. However, rNOE imaging is time-consuming and prone to biases stemming from the water T1 and the semisolid magnetization transfer (MT) contrasts. Here, we developed a rapid rNOE quantification approach, combining magnetic resonance fingerprinting (MRF) acquisition with deep-learning-based reconstruction. The method was systematically validated using tissue-mimicking phantoms, wild-type mice (n = 7), and healthy human volunteers (n = 5). In vitro rNOE parameter maps generated by MRF were highly correlated with ground truth (r > 0.98, p < 0.001). Simultaneous mapping of the rNOE and the semisolid MT exchange parameters in mice and humans were in agreement with previously reported literature values. Whole-brain 3D parameter mapping in humans took less than 5 min (282 s for acquisition and less than 2 s for reconstruction). With its demonstrated ability to rapidly extract quantitative molecular maps, deep rNOE-MRF can potentially serve as a valuable tool for the characterization and detection of molecular abnormalities in vivo.
Collapse
Affiliation(s)
- Inbal Power
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Michal Rivlin
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
- School of Chemistry, Tel Aviv University, Tel Aviv, Israel
| | - Hagar Shmuely
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Moritz Zaiss
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Gil Navon
- School of Chemistry, Tel Aviv University, Tel Aviv, Israel
| | - Or Perlman
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
17
|
Ning Y, Yuwen Zhou I, Caravan P. Quantitative in Vivo Molecular MRI. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407262. [PMID: 39279542 PMCID: PMC11530320 DOI: 10.1002/adma.202407262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/29/2024] [Indexed: 09/18/2024]
Abstract
Molecular magnetic resonance imaging (MRI) combines chemistry, chemical biology, and imaging techniques to track molecular events non-invasively. Quantitative molecular MRI aims to provide meaningful, reproducible numerical measurements of molecular processes or biochemical targets within the body. In this review, the classifications of molecular MRI probes based on their signal-generating mechanism and functionality are first described. From there, the primary considerations for in vitro characterization and in vivo validation of molecular MRI probes, including how to avoid pitfalls and biases are discussed. Then, recommendations on imaging acquisition protocols and analysis methods to establish quantitative relationships between MRI signal change induced by the probes and the molecular processes of interest are provided. Finally, several representative case studies are highlighted that incorporate these features. Quantitative molecular MRI is a multidisciplinary research area incorporating expertise in chemical biology, inorganic chemistry, molecular probes, imaging physics, drug development, pathobiology, and medicine. The purpose of this review is to provide guidance to chemists developing MR imaging probes and methods in terms of in vitro and in vivo validation to accelerate the translation of these new quantitative tools for non-invasive imaging of biological processes.
Collapse
Affiliation(s)
- Yingying Ning
- Spin-X Institute, School of Chemistry and Chemical Engineering, School of Biomedical Sciences and Engineering, State Key Laboratory of Luminescent Materials and Devices, Guangdong-Hong Kong-Macao Joint Laboratory of Optoelectronic and Magnetic Functional Materials, South China University of Technology, Guangzhou 510641, China
| | - Iris Yuwen Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| |
Collapse
|
18
|
Yu FF, Rathnakar J, Ryder B, Hitt B, Kashmer OM, Sherry AD, Vinogradov E. Differentiation and characterization of healthy versus pathological tau using chemical exchange saturation transfer. NMR IN BIOMEDICINE 2024; 37:e5160. [PMID: 38646677 DOI: 10.1002/nbm.5160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 02/29/2024] [Accepted: 03/17/2024] [Indexed: 04/23/2024]
Abstract
Neurofibrillary tangles of tau constitute one of the key biological hallmarks of Alzheimer's disease. Currently, the assessment of regional tau accumulation requires intravenous administration of radioactive tracers for PET imaging. A noninvasive MRI-based solution would have significant clinical implications. Herein, we utilized an MRI technique known as chemical exchange saturation transfer (CEST) to determine the imaging signature of tau in both its monomeric and pathologic fibrillated conformations. Three sets of purified recombinant full-length (4R) tau protein were prepared for collection of CEST spectra using a 9.4 T NMR spectrometer at varying temperatures (25, 37, and 42 °C) and RF intensities (0.7, 1.0, 1.5, and 2.2 μT). Monomeric and fibrillated tau were readily distinguished based on their Z-spectrum profiles. Fibrillated tau demonstrated a less prominent peak at 3.5 ppm with additional peaks near 0.5 and 1.5 ppm. No significant differences were identified between fibrillated tau prepared using heparin versus seed-competent tau. In conclusion, monomeric and fibrillated tau can be readily detected and distinguished based on their CEST-derived Z-spectra, pointing to the potential utility of CEST-MRI as a noninvasive biomarker of regional pathologic tau accumulation in the brain. Further testing and validation in vitro and in vivo will be necessary before this can be applied clinically.
Collapse
Affiliation(s)
- Fang Frank Yu
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - James Rathnakar
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bryan Ryder
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Brian Hitt
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurology, UCI Medical Center, Orange, California, USA
| | - Omar M Kashmer
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - A Dean Sherry
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Chemistry & Biochemistry, University of Texas at Dallas, Dallas, Texas, USA
| | - Elena Vinogradov
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
19
|
Wu Q, Gong P, Liu S, Li Y, Liang D, Zheng H, Wu Y. B 1 inhomogeneity corrected CEST MRI based on direct saturation removed omega plot model at 5T. Magn Reson Med 2024; 92:532-542. [PMID: 38650080 DOI: 10.1002/mrm.30112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/23/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024]
Abstract
PURPOSE CEST can image macromolecules/compounds via detecting chemical exchange between labile protons and bulk water. B1 field inhomogeneity impairs CEST quantification. Conventional B1 inhomogeneity correction methods depend on interpolation algorithms, B1 choices, acquisition number or calibration curves, making reliable correction challenging. This study proposed a novel B1 inhomogeneity correction method based on a direct saturation (DS) removed omega plot model. METHODS Four healthy volunteers underwent B1 field mapping and CEST imaging under four nominal B1 levels of 0.75, 1.0, 1.5, and 2.0 μT at 5T. DS was resolved using a multi-pool Lorentzian model and removed from respective Z spectrum. Residual spectral signals were used to construct the omega plot as a linear function of 1/B 1 2 $$ {B}_1^2 $$ , from which corrected signals at nominal B1 levels were calculated. Routine asymmetry analysis was conducted to quantify amide proton transfer (APT) effect. Its distribution across white matter was compared before and after B1 inhomogeneity correction and also with the conventional interpolation approach. RESULTS B1 inhomogeneity yielded conspicuous artifact on APT images. Such artifact was mitigated by the proposed method. Homogeneous APT maps were shown with SD consistently smaller than that before B1 inhomogeneity correction and the interpolation method. Moreover, B1 inhomogeneity correction from two and four CEST acquisitions yielded similar results, superior over the interpolation method that derived inconsistent APT contrasts among different B1 choices. CONCLUSION The proposed method enables reliable B1 inhomogeneity correction from at least two CEST acquisitions, providing an effective way to improve quantitative CEST MRI.
Collapse
Affiliation(s)
- Qiting Wu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Pengcheng Gong
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Department of Biomedical Engineering, Chongqing University of Technology, Chongqing, China
| | - Shengping Liu
- Department of Biomedical Engineering, Chongqing University of Technology, Chongqing, China
| | - Ye Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Dong Liang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yin Wu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
20
|
Pemmasani Prabakaran RS, Park SW, Lai JHC, Wang K, Xu J, Chen Z, Ilyas AMO, Liu H, Huang J, Chan KWY. Deep-learning-based super-resolution for accelerating chemical exchange saturation transfer MRI. NMR IN BIOMEDICINE 2024; 37:e5130. [PMID: 38491754 DOI: 10.1002/nbm.5130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 03/18/2024]
Abstract
Chemical exchange saturation transfer (CEST) MRI is a molecular imaging tool that provides physiological information about tissues, making it an invaluable tool for disease diagnosis and guided treatment. Its clinical application requires the acquisition of high-resolution images capable of accurately identifying subtle regional changes in vivo, while simultaneously maintaining a high level of spectral resolution. However, the acquisition of such high-resolution images is time consuming, presenting a challenge for practical implementation in clinical settings. Among several techniques that have been explored to reduce the acquisition time in MRI, deep-learning-based super-resolution (DLSR) is a promising approach to address this problem due to its adaptability to any acquisition sequence and hardware. However, its translation to CEST MRI has been hindered by the lack of the large CEST datasets required for network development. Thus, we aim to develop a DLSR method, named DLSR-CEST, to reduce the acquisition time for CEST MRI by reconstructing high-resolution images from fast low-resolution acquisitions. This is achieved by first pretraining the DLSR-CEST on human brain T1w and T2w images to initialize the weights of the network and then training the network on very small human and mouse brain CEST datasets to fine-tune the weights. Using the trained DLSR-CEST network, the reconstructed CEST source images exhibited improved spatial resolution in both peak signal-to-noise ratio and structural similarity index measure metrics at all downsampling factors (2-8). Moreover, amide CEST and relayed nuclear Overhauser effect maps extrapolated from the DLSR-CEST source images exhibited high spatial resolution and low normalized root mean square error, indicating a negligible loss in Z-spectrum information. Therefore, our DLSR-CEST demonstrated a robust reconstruction of high-resolution CEST source images from fast low-resolution acquisitions, thereby improving the spatial resolution and preserving most Z-spectrum information.
Collapse
Affiliation(s)
- Rohith Saai Pemmasani Prabakaran
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong, China
| | - Se Weon Park
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong, China
| | - Joseph H C Lai
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Kexin Wang
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, Maryland, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jiadi Xu
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, Maryland, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zilin Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | | | - Huabing Liu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Jianpan Huang
- Department of Diagnostic Radiology, The University of Hong Kong, Hong Kong, China
| | - Kannie W Y Chan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong, China
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Tung Biomedical Sciences Centre, Hong Kong, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
21
|
Lam B, Velasquez M, Ogiyama T, Godines K, Szu FY, Velasquez-Mao AJ, AlGhuraibawi W, Wang J, Messersmith PB, Vandsburger MH. Imaging of adeno-associated viral capsids for purposes of gene editing using CEST NMR/MRI. Magn Reson Med 2024; 92:792-806. [PMID: 38651648 PMCID: PMC11142879 DOI: 10.1002/mrm.30058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/19/2024] [Accepted: 02/03/2024] [Indexed: 04/25/2024]
Abstract
PURPOSE Gene therapy using adeno-associated virus (AAV) vector-mediated gene delivery has undergone substantial growth in recent years with promising results in both preclinical and clinical studies, as well as emerging regulatory approval. However, the inability to quantify the efficacy of gene therapy from cellular delivery of gene-editing technology to specific functional outcomes is an obstacle for efficient development of gene therapy treatments. Building on prior works that used the CEST reporter gene lysine rich protein, we hypothesized that AAV viral capsids may generate endogenous CEST contrast from an abundance of surface lysine residues. METHODS NMR experiments were performed on isolated solutions of AAV serotypes 1-9 on a Bruker 800-MHz vertical scanner. In vitro experiments were performed for testing of CEST-NMR contrast of AAV2 capsids under varying pH, density, biological transduction stage, and across multiple serotypes and mixed biological media. Reverse transcriptase-polymerase chain reaction was used to quantify virus concentration. Subsequent experiments at 7 T optimized CEST saturation schemes for AAV contrast detection and detected AAV2 particles encapsulated in a biocompatible hydrogel administered in the hind limb of mice. RESULTS CEST-NMR experiments revealed CEST contrast up to 52% for AAV2 viral capsids between 0.6 and 0.8 ppm. CEST contrast generated by AAV2 demonstrated high levels of CEST contrast across a variety of chemical environments, concentrations, and saturation schemes. AAV2 CEST contrast displayed significant positive correlations with capsid density (R2 > 0.99, p < 0.001), pH (R2 = 0.97, p = 0.01), and viral titer per cell count (R2 = 0.92, p < 0.001). Transition to a preclinical field strength yielded up to 11.8% CEST contrast following optimization of saturation parameters. In vivo detection revealed statistically significant molecular contrast between viral and empty hydrogels using both mean values (4.67 ± 0.75% AAV2 vs. 3.47 ± 0.87% empty hydrogel, p = 0.02) and quantile analysis. CONCLUSION AAV2 viral capsids exhibit strong capacity as an endogenous CEST contrast agent and can potentially be used for monitoring and evaluation of AAV vector-mediated gene therapy protocols.
Collapse
Affiliation(s)
- Bonnie Lam
- Department of Bioengineering, UC Berkeley, Berkeley, California, USA
| | - Mark Velasquez
- Department of Bioengineering, UC Berkeley, Berkeley, California, USA
| | - Tomoko Ogiyama
- Department of Bioengineering, UC Berkeley, Berkeley, California, USA
| | - Kevin Godines
- Department of Bioengineering, UC Berkeley, Berkeley, California, USA
| | - Fan-Yun Szu
- Department of Bioengineering, UC Berkeley, Berkeley, California, USA
| | - A J Velasquez-Mao
- Department of Bioengineering, UC Berkeley, Berkeley, California, USA
| | | | - Jingshen Wang
- Division of Biostatistics, UC Berkeley, Berkeley, California, USA
| | - Phillip B Messersmith
- Department of Bioengineering, UC Berkeley, Berkeley, California, USA
- Department of Materials Science and Engineering, UC Berkeley, Berkeley, California, USA
- Materials Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | | |
Collapse
|
22
|
Liu C, Li Z, Chen Z, Zhao B, Zheng Z, Song X. Highly-accelerated CEST MRI using frequency-offset-dependent k-space sampling and deep-learning reconstruction. Magn Reson Med 2024; 92:688-701. [PMID: 38623899 DOI: 10.1002/mrm.30089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/31/2024] [Accepted: 03/02/2024] [Indexed: 04/17/2024]
Abstract
PURPOSE To develop a highly accelerated CEST Z-spectral acquisition method using a specifically-designed k-space sampling pattern and corresponding deep-learning-based reconstruction. METHODS For k-space down-sampling, a customized pattern was proposed for CEST, with the randomized probability following a frequency-offset-dependent (FOD) function in the direction of saturation offset. For reconstruction, the convolution network (CNN) was enhanced with a Partially Separable (PS) function to optimize the spatial domain and frequency domain separately. Retrospective experiments on a self-acquired human brain dataset (13 healthy adults and 15 brain tumor patients) were conducted using k-space resampling. The prospective performance was also assessed on six healthy subjects. RESULTS In retrospective experiments, the combination of FOD sampling and PS network (FOD + PSN) showed the best quantitative metrics for reconstruction, outperforming three other combinations of conventional sampling with varying density and a regular CNN (nMSE and SSIM, p < 0.001 for healthy subjects). Across all acceleration factors from 4 to 14, the FOD + PSN approach consistently outperformed the comparative methods in four contrast maps including MTRasym, MTRrex, as well as the Lorentzian Difference maps of amide and nuclear Overhauser effect (NOE). In the subspace replacement experiment, the error distribution demonstrated the denoising benefits achieved in the spatial subspace. Finally, our prospective results obtained from healthy adults and brain tumor patients (14×) exhibited the initial feasibility of our method, albeit with less accurate reconstruction than retrospective ones. CONCLUSION The combination of FOD sampling and PSN reconstruction enabled highly accelerated CEST MRI acquisition, which may facilitate CEST metabolic MRI for brain tumor patients.
Collapse
Affiliation(s)
- Chuyu Liu
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, Tsinghua University, Beijing, China
| | - Zhongsen Li
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, Tsinghua University, Beijing, China
| | - Zhensen Chen
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
| | - Benqi Zhao
- Department of Radiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Zhuozhao Zheng
- Department of Radiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Xiaolei Song
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, Tsinghua University, Beijing, China
| |
Collapse
|
23
|
Gammaraccio F, Villano D, Irrera P, Anemone AA, Carella A, Corrado A, Longo DL. Development and Validation of Four Different Methods to Improve MRI-CEST Tumor pH Mapping in Presence of Fat. J Imaging 2024; 10:166. [PMID: 39057737 PMCID: PMC11277679 DOI: 10.3390/jimaging10070166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/28/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
CEST-MRI is an emerging imaging technique suitable for various in vivo applications, including the quantification of tumor acidosis. Traditionally, CEST contrast is calculated by asymmetry analysis, but the presence of fat signals leads to wrong contrast quantification and hence to inaccurate pH measurements. In this study, we investigated four post-processing approaches to overcome fat signal influences and enable correct CEST contrast calculations and tumor pH measurements using iopamidol. The proposed methods involve replacing the Z-spectrum region affected by fat peaks by (i) using a linear interpolation of the fat frequencies, (ii) applying water pool Lorentzian fitting, (iii) considering only the positive part of the Z-spectrum, or (iv) calculating a correction factor for the ratiometric value. In vitro and in vivo studies demonstrated the possibility of using these approaches to calculate CEST contrast and then to measure tumor pH, even in the presence of moderate to high fat fraction values. However, only the method based on the water pool Lorentzian fitting produced highly accurate results in terms of pH measurement in tumor-bearing mice with low and high fat contents.
Collapse
Affiliation(s)
- Francesco Gammaraccio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Torino, Italy
| | - Daisy Villano
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Torino, Italy
| | - Pietro Irrera
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), 10126 Torino, Italy
| | - Annasofia A. Anemone
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Torino, Italy
| | - Antonella Carella
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), 10126 Torino, Italy
| | - Alessia Corrado
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), 10126 Torino, Italy
| | - Dario Livio Longo
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), 10126 Torino, Italy
| |
Collapse
|
24
|
Rosa E, Di Gregorio E, Ferrauto G, Diaferia C, Gallo E, Terreno E, Accardo A. Hybrid PNA-peptide hydrogels as injectable CEST-MRI agents. J Mater Chem B 2024; 12:6371-6383. [PMID: 38864345 DOI: 10.1039/d4tb00358f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
The self-assembly of peptides and peptide analogues may be exploited to develop platforms for different biomedical applications, among which CEST-MRI (chemical exchange saturation transfer magnetic resonance imaging) represents one of the most attractive techniques to be explored as a novel metal-free contrast approach in imaging acquisitions. A lysine-containing peptide sequence (LIVAGK-NH2, named K2) was thus modified by insertion, at the N-terminus, of a peptide nucleic acid (PNA) base, leading to a primary amine suitable for the signal generation. a-K2, c-K2, g-K2 and t-K2 peptides were synthesized and characterized. The c-K2 sequence displayed gelling properties and the Watson and Crick pairing, arising from its combination with g-K2, allowed a significant increase in the mechanical responsivity of the hydrogel. These matrices were able to generate a CEST signal around 2.5 ppm from water and, after assessing their cytocompatibility on GL261 (murine glioma), TS/a (murine breast carcinoma), and 3T3-NIH (murine fibroblasts) cell lines, their capability to work as implants for in vivo detection, was proved by intratumor injection in Balb/c mice inoculated with TS/a murine breast cancer cells.
Collapse
Affiliation(s)
- Elisabetta Rosa
- Department of Pharmacy and Interuniversity Research Centre on Bioactive Peptides (CIRPeB) "Carlo Pedone", University of Naples "Federico II", Via D. Montesano 49, Naples 80131, Italy.
| | - Enza Di Gregorio
- Molecular and Preclinical Imaging Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, Turin, Italy.
| | - Giuseppe Ferrauto
- Molecular and Preclinical Imaging Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, Turin, Italy.
| | - Carlo Diaferia
- Department of Pharmacy and Interuniversity Research Centre on Bioactive Peptides (CIRPeB) "Carlo Pedone", University of Naples "Federico II", Via D. Montesano 49, Naples 80131, Italy.
| | - Enrico Gallo
- IRCCS Synlab SDN, Via Gianturco 113, Naples, 80143, Italy
| | - Enzo Terreno
- Molecular and Preclinical Imaging Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, Turin, Italy.
| | - Antonella Accardo
- Department of Pharmacy and Interuniversity Research Centre on Bioactive Peptides (CIRPeB) "Carlo Pedone", University of Naples "Federico II", Via D. Montesano 49, Naples 80131, Italy.
| |
Collapse
|
25
|
Mohanta Z, Gori S, McMahon MT. Intramolecular Hydrogen Bonding Based CEST MRI Contrast Agents As an Emerging Design Strategy: A Mini-Review. ACS OMEGA 2024; 9:27755-27765. [PMID: 38973929 PMCID: PMC11223143 DOI: 10.1021/acsomega.4c02296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/21/2024] [Accepted: 06/05/2024] [Indexed: 07/09/2024]
Abstract
Intramolecular hydrogen bonding-based chemical exchange saturation transfer magnetic resonance imaging (CEST MRI) contrast agents represent an innovative design strategy aiming to overcome limitations in diamagnetic CEST (diaCEST) MRI contrast agent specificity and also those associated with traditional metal-based MRI contrast agents. Ward and Balaban's proposal of small diamagnetic compounds marked a paradigm shift in contrast-based radiologic research, inspiring extensive investigations since 2000. These contrast agents leverage labile hydrogen bonds, serving as chemical exchange sites to induce saturation of water. The selective manipulation of radiofrequency (RF) allows for optimized signal contrast in soft tissue, with a significant signal amplification even at low probe concentrations, mitigating concerns about dose-dependent toxicities. This mini-review delves into the evolution of CEST MRI, its classification, and the strategic design principles of synthetic small molecules containing intramolecular hydrogen bonds. With a focus on applications and potential clinical relevance, the authors highlight the promising role of intramolecular hydrogen bonding-based CEST MRI in diverse medical contexts, especially renal imaging and pH mapping, paving the way for enhanced molecular imaging capabilities. Ongoing research endeavors aim to further optimize and expand the utility of these contrast agents, underscoring their transformative potential in clinical diagnostics and imaging.
Collapse
Affiliation(s)
- Zinia Mohanta
- Russell
H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- F.M.
Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, Maryland 21205, United States
| | - Sadakatali Gori
- Center
for Translational Pharmacology, Department of Pharmacy and Pharmaceutical
Sciences, St. Jude Children’s Research
Hospital, Memphis, Tennessee 38105-3678, United States
| | - Michael T. McMahon
- Russell
H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- F.M.
Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, Maryland 21205, United States
| |
Collapse
|
26
|
Mohanta Z, Stabinska J, Gilad AA, Barker PB, McMahon MT. The Proton Resonance Enhancement for CEST imaging and Shift Exchange (PRECISE) family of RF pulse shapes for Chemical Exchange Saturation Transfer MRI. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.19.599565. [PMID: 38948741 PMCID: PMC11212941 DOI: 10.1101/2024.06.19.599565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Purpose To optimize a 100 msec pulse for producing CEST MRI contrast and evaluate in mice. Methods A gradient ascent algorithm was employed to generate a family of 100 point, 100 msec pulses for use in CEST pulse trains ('PRECISE'). Gradient ascent optimizations were performed for exchange rates (k ca ) = 500 s -1 , 1,500 s -1 , 2,500 s -1 , 3,500 s -1 and 4,500 s -1 and offsets (Δω) = 9.6, 7.8, 4.2 and 2.0 ppm. 7 PRECISE pulse shapes were tested on an 11.7 T scanner using a phantom containing three representative CEST agents with peak saturation B 1 = 4 μT. The pulse producing the most contrast in phantoms was then evaluated for CEST MRI pH mapping of the kidneys in healthy mice after iopamidol administration. Results The most promising pulse in terms of contrast performance across all three phantoms was the 9.6 ppm, 2500 s -1 optimized pulse with ∼2.7 x improvement over Gaussian and ∼1.3x's over Fermi pulses. This pulse also displayed a large improvement in contrast over the Gaussian pulse after administration of iopamidol in live mice. Conclusion A new 100 msec pulse was developed based on gradient ascent optimizations which produced better contrast compared to standard Gaussian and Fermi pulses in phantoms. This shape also showed a substantial improvement for CEST MRI pH mapping in live mice over the Gaussian shape and appears promising for a wide range of CEST applications.
Collapse
|
27
|
Shaghaghi M, Damen FC, Li W, Tai LM, Cai K. Induced saturation transfer recovery steady states (iSTRESS) for proton exchange rate mapping with CEST MRI, a preliminary study. Magn Reson Imaging 2024; 109:264-270. [PMID: 38522624 PMCID: PMC11440908 DOI: 10.1016/j.mri.2024.03.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/21/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
Proton exchange underpins essential mechanisms in diverse MR imaging contrasts. Omega plots have proven effective in mapping proton exchange rates (kex) in live human brains, enabling the differentiation of MS lesion activities and characterization of ischemic stroke. However, Omega plots require extended saturation durations (typically 5 to 10 s), resulting in high specific absorption rates (SAR) that can hinder clinical feasibility. In this study, we introduce a novel kex mapping approach, named induced Saturation Transfer Recovery Steady-States (iSTRESS). iSTRESS integrates an excitation flip angle pulse prior to chemical exchange saturation transfer (CEST) saturation, effectively aligning the magnetization with its steady-state value. This innovation reduces saturation times and mitigates SAR concerns. The formula for iSTRESS-based kex quantification was derived theoretically, involving two measurements with distinct excitation flip angles and saturation B1 values. Bloch-McConnell simulations confirmed that iSTRESS-based kex values closely matched input values (R2 > 0.99). An iSTRESS MRI sequence was implemented on a 9.4 T preclinical MRI, imaging protein phantoms with pH values ranging from 6.2 to 7.4 (n = 4). Z-spectra were acquired using excitation flip angles of 30° and 60°, followed by CEST saturation at powers of 30 and 120 Hz respectively, with a total saturation time of <1 s, resulting in two iSTRESS states for kex mapping. kex maps derived from the phantom study exhibited a linear correlation (R2 > 0.99) with Omega plot results. The developed iSTRESS method allows for kex quantification with significantly reduced saturation times, effectively minimizing SAR concerns.
Collapse
Affiliation(s)
- Mehran Shaghaghi
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Frederick C Damen
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Weiguo Li
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Kejia Cai
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA.
| |
Collapse
|
28
|
Yablonskiy DA, Sukstanskii AL. Quantum dipole interactions and transient hydrogen bond orientation order in cells, cellular membranes and myelin sheath: Implications for MRI signal relaxation, anisotropy, and T 1 magnetic field dependence. Magn Reson Med 2024; 91:2597-2611. [PMID: 38241135 PMCID: PMC10997466 DOI: 10.1002/mrm.29996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 01/21/2024]
Abstract
PURPOSE Despite significant impact on the study of human brain, MRI lacks a theory of signal formation that integrates quantum interactions involving proton dipoles (a primary MRI signal source) with brain intricate cellular environment. The purpose of the present study is developing such a theory. METHODS We introduce the Transient Hydrogen Bond (THB) model, where THB-mediated quantum dipole interactions between water and protons of hydrophilic heads of amphipathic biomolecules forming cells, cellular membranes and myelin sheath serve as a major source of MR signal relaxation. RESULTS The THB theory predicts the existence of a hydrogen-bond-driven structural order of dipole-dipole connections within THBs as a primary factor for the anisotropy observed in MRI signal relaxation. We have also demonstrated that the conventional Lorentzian spectral density function decreases too fast at high frequencies to adequately capture the field dependence of brain MRI signal relaxation. To bridge this gap, we introduced a stretched spectral density function that surpasses the limitations of Lorentzian dispersion. In human brain, our findings reveal that at any time point only about 4% to 7% of water protons are engaged in quantum encounters within THBs. These ultra-short (2 to 3 ns), but frequent quantum spin exchanges lead to gradual recovery of magnetization toward thermodynamic equilibrium, that is, relaxation of MRI signal. CONCLUSION By incorporating quantum proton interactions involved in brain imaging, the THB approach introduces new insights on the complex relationship between brain tissue cellular structure and MRI measurements, thus offering a promising new tool for better understanding of brain microstructure in health and disease.
Collapse
Affiliation(s)
- Dmitriy A. Yablonskiy
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, 4525 Scott Ave. Room 3216, St. Louis MO, 63110
- Hope Center for Neurological Disorder, 660 S. Euclid Ave., St. Louis, Missouri 63110
- Knight Alzheimer Disease Research Center, 4488 Forest Park Ave., St. Louis, MO 63108
- Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Drive, St. Louis, MO 63130
| | - Alexander L. Sukstanskii
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, 4525 Scott Ave. Room 3216, St. Louis MO, 63110
| |
Collapse
|
29
|
Sheng L, Yuan E, Yuan F, Song B. Amide proton transfer-weighted imaging of the abdomen: Current progress and future directions. Magn Reson Imaging 2024; 107:88-99. [PMID: 38242255 DOI: 10.1016/j.mri.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/13/2024] [Accepted: 01/14/2024] [Indexed: 01/21/2024]
Abstract
The chemical exchange saturation transfer technique serves as a valuable tool for generating in vivo image contrast based on the content of various proton groups, including amide protons, amine protons, and aliphatic protons. Among these, amide proton transfer-weighted (APTw) imaging has seen extensive development as a means to assess the biochemical status of lesions. The exchange from saturated amide protons to bulk water protons during and following the saturation ratio frequency pulse contributes to detectable APT signals. While APTw imaging has garnered significant attention in the central nervous system, demonstrating noteworthy findings in cerebral neoplasia, stroke, and Alzheimer's disease over the past decade, its application in the abdomen has been a relatively recent progression. Notably, studies have explored its utility in hepatocellular carcinoma, prostate cancer, and cervical carcinoma within the abdominal context. Despite these advancements, there is a paucity of reviews on APTw imaging in abdominal applications. This paper aims to fill this gap by providing a concise overview of the fundamental theories underpinning APTw imaging. Additionally, we systematically summarize its diverse clinical applications in the abdomen, with a particular focus on the digestive and urogenital systems. Finally, the manuscript concludes by discussing technical limitations and factors influencing APTw imaging in abdominal applications, along with prospects for future research.
Collapse
Affiliation(s)
- Liuji Sheng
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Enyu Yuan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fang Yuan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Radiology, Sanya People's Hospital, Sanya, Hainan, China.
| |
Collapse
|
30
|
Di Gregorio E, Papi C, Conti L, Di Lorenzo A, Cavallari E, Salvatore M, Cavaliere C, Ferrauto G, Aime S. A Magnetic Resonance Imaging-Chemical Exchange Saturation Transfer (MRI-CEST) Method for the Detection of Water Cycling across Cellular Membranes. Angew Chem Int Ed Engl 2024; 63:e202313485. [PMID: 37905585 DOI: 10.1002/anie.202313485] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/02/2023]
Abstract
Water cycling across the membrane transporters is considered a hallmark of cellular metabolism and it could be of high diagnostic relevance in the characterization of tumors and other diseases. The method relies on the response of intracellular proton exchanging molecules to the presence of extracellular Gd-based contrast agents (GBCAs). Paramagnetic GBCAs enhances the relaxation rate of water molecules in the extracellular compartment and, through membrane exchange, the relaxation enhancement is transferred to intracellular molecules. The effect is detected at the MRI-CEST (Magnetic Resonance Imaging - Chemical Exchange Saturation Transfer) signal of intracellular proton exchanging molecules. The magnitude of the change in the CEST response reports on water cycling across the membrane. The method has been tested on Red Blood Cells and on orthotopic murine models of breast cancer with different degree of malignancy (4T1, TS/A and 168FARN). The distribution of voxels reporting on membrane permeability fits well with the cells' aggressiveness and acts as an early reporter to monitor therapeutic treatments.
Collapse
Affiliation(s)
- Enza Di Gregorio
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Chiara Papi
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Laura Conti
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Antonino Di Lorenzo
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Eleonora Cavallari
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Marco Salvatore
- IRCCS SDN SynLab, Via E. Gianturco 113, 80143, Napoli, Italy
| | - Carlo Cavaliere
- IRCCS SDN SynLab, Via E. Gianturco 113, 80143, Napoli, Italy
| | - Giuseppe Ferrauto
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Silvio Aime
- IRCCS SDN SynLab, Via E. Gianturco 113, 80143, Napoli, Italy
| |
Collapse
|
31
|
Zhang Z. Editorial for "A Pilot Study of Ratiometric Creatine CEST MRI Assessment of Rabbit Skeletal Muscle Energy Metabolism at 3 T". J Magn Reson Imaging 2024; 59:209-210. [PMID: 37317058 DOI: 10.1002/jmri.28871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/16/2023] Open
Affiliation(s)
- Zhongwei Zhang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
32
|
Wu Q, Qi Y, Gong P, Huang B, Cheng G, Liang D, Zheng H, Sun PZ, Wu Y. Fast and robust pulsed chemical exchange saturation transfer (CEST) MRI using a quasi-steady-state (QUASS) algorithm at 3 T. Magn Reson Imaging 2024; 105:29-36. [PMID: 37898416 DOI: 10.1016/j.mri.2023.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/21/2023] [Accepted: 10/22/2023] [Indexed: 10/30/2023]
Abstract
Chemical exchange saturation transfer (CEST) has emerged as a powerful technique to image dilute labile protons. However, its measurement depends on the RF saturation duration (Tsat) and relaxation delay (Trec). Although the recently developed quasi-steady-state (QUASS) solution can reconstruct equilibrium CEST effects under continuous-wave RF saturation, it does not apply to pulsed-CEST MRI on clinical scanners with restricted hardware or specific absorption rate limits. This study proposed a QUASS algorithm for pulsed-CEST MRI and evaluated its performance in muscle CEST measurement. An approximated expression of a steady-state pulsed-CEST signal was incorporated in the off-resonance spin-lock model, from which the QUASS pulsed-CEST effect was derived. Numerical simulation, creatine phantom, and healthy volunteer scans were conducted at 3 T. The CEST effect was quantified with asymmetry analysis in the simulation and phantom experiments. CEST effects of creatine, amide proton transfer, phosphocreatine, and combined magnetization transfer and nuclear Overhauser effects were isolated from a multi-pool Lorentzian model in muscles. Apparent and QUASS CEST measurements were compared under different Tsat/Trec and duty cycles. Paired Student's t-test was employed with P < 0.05 as statistically significant. The simulation, phantom, and human studies showed the strong impact of Tsat/Trec on apparent CEST measurements, which were significantly smaller than the corresponding QUASS CEST measures, especially under short Tsat/Trec times. In comparison, the QUASS algorithm mitigates such impact and enables accurate CEST measurements under short Tsat/Trec times. In conclusion, the QUASS algorithm can accelerate robust pulsed-CEST MRI, promising the efficient detection and evaluation of muscle diseases in clinical settings.
Collapse
Affiliation(s)
- Qiting Wu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China; Medical AI Lab, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Yulong Qi
- Department of Medical Imaging, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Pengcheng Gong
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Bingsheng Huang
- Medical AI Lab, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Guanxun Cheng
- Department of Medical Imaging, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Dong Liang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Phillip Zhe Sun
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Yin Wu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China.
| |
Collapse
|
33
|
Zhuang C, Chen B, Wu S, Xu L, Zhang X, Zheng X, Chen Y, Geng Y, Guan J, Lin Y, Wilman AH, Wu R. Repurposing of the PET Probe Prototype PiB for Label and Radiation-Free CEST MRI Molecular Imaging of Amyloid. ACS Chem Neurosci 2023; 14:4344-4351. [PMID: 38061891 PMCID: PMC10741654 DOI: 10.1021/acschemneuro.3c00539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/21/2023] Open
Abstract
Positron emission tomography (PET) probes are specific and sensitive while suffering from radiation risk. It is worthwhile to explore the chemical emission saturation transfer (CEST) effects of the probe prototypes and repurpose them for CEST imaging to avoid radiation. In this study, we used 11C-PiB as an example of a PET probe for detecting amyloid and tested the feasibility of repurposing this PET probe prototype, PiB, for CEST imaging. After optimizing the parameters through preliminary phantom experiments, we used APP/PS1 transgenic mice and age-matched C57 mice for in vivo CEST magnetic resonance imaging (MRI) of amyloid. Furthermore, the pathological assessment was conducted on the same brain slices to evaluate the correlation between the CEST MRI signal abnormality and β-amyloid deposition detected by immunohistochemical staining. In our results, the Z-spectra revealed an apparent CEST effect that peaked at approximately 6 ppm. APP/PS1 mice as young as 9 months injected with PiB showed a significantly higher CEST effect compared to the control groups. The hyperintense region was correlated with the Aβ deposition shown by pathological staining. In conclusion, repurposing the PET probe prototype for CEST MRI imaging is feasible and enables label- and radiation-free detection of the amyloid while maintaining the sensitivity and specificity of the ligand. This study opens the door to developing CEST probes based on PET probe prototypes.
Collapse
Affiliation(s)
- Caiyu Zhuang
- Department
of Radiology, Second Affiliated Hospital, Shantou University Medical College, Shantou 515041, China
- Department
of Radiology, First Affiliated Hospital, Shantou University Medical College, Shantou 515041, China
| | - Beibei Chen
- Department
of Radiology, Second Affiliated Hospital, Shantou University Medical College, Shantou 515041, China
| | - Shuohua Wu
- Department
of Radiology, Second Affiliated Hospital, Shantou University Medical College, Shantou 515041, China
| | - Liang Xu
- Department
of Medical Imaging, Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Xiaolei Zhang
- Department
of Radiology, Second Affiliated Hospital, Shantou University Medical College, Shantou 515041, China
| | - Xinhui Zheng
- Department
of Radiology, Second Affiliated Hospital, Shantou University Medical College, Shantou 515041, China
| | - Yue Chen
- Department
of Radiology, Second Affiliated Hospital, Shantou University Medical College, Shantou 515041, China
| | - Yiqun Geng
- Laboratory
of Molecular Pathology, Shantou University
Medical College, Shantou 515041, China
| | - Jitian Guan
- Department
of Radiology, Second Affiliated Hospital, Shantou University Medical College, Shantou 515041, China
| | - Yan Lin
- Department
of Radiology, Second Affiliated Hospital, Shantou University Medical College, Shantou 515041, China
- Provincial
Key Laboratory for Breast Cancer Diagnosis and Treatment, Shantou 515041, China
| | - Alan H. Wilman
- The Department
of Biomedical Engineering, University of
Alberta, Edmonton, Alberta T6G 1H9, Canada
| | - Renhua Wu
- Department
of Radiology, Second Affiliated Hospital, Shantou University Medical College, Shantou 515041, China
- Provincial
Key Laboratory for Breast Cancer Diagnosis and Treatment, Shantou 515041, China
| |
Collapse
|
34
|
Ali SM, Yadav NN, Wirestam R, Singh M, Heo HY, van Zijl PC, Knutsson L. Deep learning-based Lorentzian fitting of water saturation shift referencing spectra in MRI. Magn Reson Med 2023; 90:1610-1624. [PMID: 37279008 PMCID: PMC10524193 DOI: 10.1002/mrm.29718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/27/2023] [Accepted: 05/12/2023] [Indexed: 06/07/2023]
Abstract
PURPOSE Water saturation shift referencing (WASSR) Z-spectra are used commonly for field referencing in chemical exchange saturation transfer (CEST) MRI. However, their analysis using least-squares (LS) Lorentzian fitting is time-consuming and prone to errors because of the unavoidable noise in vivo. A deep learning-based single Lorentzian Fitting Network (sLoFNet) is proposed to overcome these shortcomings. METHODS A neural network architecture was constructed and its hyperparameters optimized. Training was conducted on a simulated and in vivo-paired data sets of discrete signal values and their corresponding Lorentzian shape parameters. The sLoFNet performance was compared with LS on several WASSR data sets (both simulated and in vivo 3T brain scans). Prediction errors, robustness against noise, effects of sampling density, and time consumption were compared. RESULTS LS and sLoFNet performed comparably in terms of RMS error and mean absolute error on all in vivo data with no statistically significant difference. Although the LS method fitted well on samples with low noise, its error increased rapidly when increasing sample noise up to 4.5%, whereas the error of sLoFNet increased only marginally. With the reduction of Z-spectral sampling density, prediction errors increased for both methods, but the increase occurred earlier (at 25 vs. 15 frequency points) and was more pronounced for LS. Furthermore, sLoFNet performed, on average, 70 times faster than the LS-method. CONCLUSION Comparisons between LS and sLoFNet on simulated and in vivo WASSR MRI Z-spectra in terms of robustness against noise and decreased sample resolution, as well as time consumption, showed significant advantages for sLoFNet.
Collapse
Affiliation(s)
| | - Nirbhay N. Yadav
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Ronnie Wirestam
- Department of Medical Radiation Physics, Lund University, Lund, Sweden
| | - Munendra Singh
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Hye-Young Heo
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Peter C. van Zijl
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Linda Knutsson
- Department of Medical Radiation Physics, Lund University, Lund, Sweden
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| |
Collapse
|
35
|
Łopuszyńska N, Węglarz WP. Contrasting Properties of Polymeric Nanocarriers for MRI-Guided Drug Delivery. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2163. [PMID: 37570481 PMCID: PMC10420849 DOI: 10.3390/nano13152163] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023]
Abstract
Poor pharmacokinetics and low aqueous solubility combined with rapid clearance from the circulation of drugs result in their limited effectiveness and generally high therapeutic doses. The use of nanocarriers for drug delivery can prevent the rapid degradation of the drug, leading to its increased half-life. It can also improve the solubility and stability of drugs, advance their distribution and targeting, ensure a sustained release, and reduce drug resistance by delivering multiple therapeutic agents simultaneously. Furthermore, nanotechnology enables the combination of therapeutics with biomedical imaging agents and other treatment modalities to overcome the challenges of disease diagnosis and therapy. Such an approach is referred to as "theranostics" and aims to offer a more patient-specific approach through the observation of the distribution of contrast agents that are linked to therapeutics. The purpose of this paper is to present the recent scientific reports on polymeric nanocarriers for MRI-guided drug delivery. Polymeric nanocarriers are a very broad and versatile group of materials for drug delivery, providing high loading capacities, improved pharmacokinetics, and biocompatibility. The main focus was on the contrasting properties of proposed polymeric nanocarriers, which can be categorized into three main groups: polymeric nanocarriers (1) with relaxation-type contrast agents, (2) with chemical exchange saturation transfer (CEST) properties, and (3) with direct detection contrast agents based on fluorinated compounds. The importance of this aspect tends to be downplayed, despite its being essential for the successful design of applicable theranostic nanocarriers for image-guided drug delivery. If available, cytotoxicity and therapeutic effects were also summarized.
Collapse
Affiliation(s)
- Natalia Łopuszyńska
- Department of Magnetic Resonance Imaging, Institute of Nuclear Physics Polish Academy of Sciences, 31-342 Cracow, Poland
| | - Władysław P. Węglarz
- Department of Magnetic Resonance Imaging, Institute of Nuclear Physics Polish Academy of Sciences, 31-342 Cracow, Poland
| |
Collapse
|
36
|
Ohno Y, Yui M, Yamamoto K, Takenaka D, Koyama H, Nagata H, Ueda T, Ikeda H, Ozawa Y, Toyama H, Yoshikawa T. Chemical Exchange Saturation Transfer MRI: Capability for Predicting Therapeutic Effect of Chemoradiotherapy on Non-Small Cell Lung Cancer Patients. J Magn Reson Imaging 2023; 58:174-186. [PMID: 36971493 DOI: 10.1002/jmri.28691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Amide proton transfer (APT) weighted chemical exchange saturation transfer CEST (APTw/CEST) magnetic resonance imaging (MRI) has been suggested as having the potential for assessing the therapeutic effect of brain tumors or rectal cancer. Moreover, diffusion-weighted imaging (DWI) and positron emission tomography fused with computed tomography by means of 2-[fluorine-18]-fluoro-2-deoxy-D-glucose (FDG-PET/CT) have been suggested as useful in same setting. PURPOSE To compare the capability of APTw/CEST imaging, DWI, and FDG-PET/CT for predicting therapeutic effect of chemoradiotherapy (CRT) on stage III non-small cell lung cancer (NSCLC) patients. STUDY TYPE Prospective. POPULATION Eighty-four consecutive patients with Stage III NSCLC, 45 men (age range, 62-75 years; mean age, 71 years) and 39 women (age range, 57-75 years; mean age, 70 years). All patients were then divided into two groups (Response Evaluation Criteria in Solid Tumors [RECIST] responders, consisting of the complete response and partial response groups, and RECIST non-responders, consisting of the stable disease and progressive disease groups). FIELD STRENGTH/SEQUENCE 3 T, echo planar imaging or fast advanced spin-echo (FASE) sequences for DWI and 2D half Fourier FASE sequences with magnetization transfer pulses for CEST imaging. ASSESSMENT Magnetization transfer ratio asymmetry (MTRasym ) at 3.5 ppm, apparent diffusion coefficient (ADC), and maximum standard uptake value (SUVmax, ) on PET/CT were assessed by means of region of interest (ROI) measurements at primary tumor. STATISTICAL TESTS Kaplan-Meier method followed by log-rank test and Cox proportional hazards regression analysis with multivariate analysis. A P value <0.05 was considered statistically significant. RESULTS Progression-free survival (PFS) and overall survival (OS) had significant difference between two groups. MTRasym at 3.5 ppm (hazard ratio [HR] = 0.70) and SUVmax (HR = 1.41) were identified as significant predictors for PFS. Tumor staging (HR = 0.57) was also significant predictors for OS. DATA CONCLUSION APTw/CEST imaging showed potential performance as DWI and FDG-PET/CT for predicting the therapeutic effect of CRT on stage III NSCLC patients. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Yoshiharu Ohno
- Department of Radiology, Fujita Health University School of Medicine, Toyoake, Japan
- Joint Research Laboratory of Advanced Medical Imaging, Fujita Health University School of Medicine, Toyoake, Japan
- Division of Functional and Diagnostic Imaging Research, Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masao Yui
- Canon Medical Systems Corporation, Otawara, Japan
| | | | - Daisuke Takenaka
- Department of Radiology, Fujita Health University School of Medicine, Toyoake, Japan
- Department of Diagnostic Radiology, Hyogo Cancer Center, Akashi, Japan
| | - Hisanobu Koyama
- Department of Radiology, Osaka Police Hospital, Osaka, Japan
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroyuki Nagata
- Joint Research Laboratory of Advanced Medical Imaging, Fujita Health University School of Medicine, Toyoake, Japan
| | - Takahiro Ueda
- Department of Radiology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Hirotaka Ikeda
- Department of Radiology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Yoshiyuki Ozawa
- Department of Radiology, Fujita Health University School of Medicine, Toyoake, Japan
- Department of Radiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroshi Toyama
- Department of Radiology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Takeshi Yoshikawa
- Department of Radiology, Fujita Health University School of Medicine, Toyoake, Japan
- Division of Functional and Diagnostic Imaging Research, Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan
- Department of Diagnostic Radiology, Hyogo Cancer Center, Akashi, Japan
| |
Collapse
|
37
|
Tian Y, Li X, Wang X, Su W, Li S, Wang W, Zhang Y, Li C, Chen M. CEST 2022-three-dimensional amide proton transfer (APT) imaging can identify the changes of cerebral cortex in Parkinson's disease. Magn Reson Imaging 2023:S0730-725X(23)00099-1. [PMID: 37356600 DOI: 10.1016/j.mri.2023.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 06/12/2023] [Indexed: 06/27/2023]
Abstract
PURPOSE Amide proton transfer (APT) imaging has shown its diagnostic and predictive superiority in PD in our previous studies using 2D APT imaging based on deep nuclei. We hypothesized that the pathophysiological abnormality of PD will change the APT-related parameters in the cerebral cortex, and the signal changes can contribute to accurate diagnosis of Parkinson's disease. METHODS 34 patients with sporadic Parkinson's disease (IPD) and 29 age- and sex-matched normal controls (NC) were enrolled in this prospective study. 3D-APT imaging and 3D-T1WI was performed in our participants. A volume-based morphometry algorithm was used and get automated cortical segmentations. Quantitative parameter maps of APT-related metrics were calculated by using SPM and MATLAB. The unpaired Student's t-test or Mann-Whitney U test was used for comparison of these values between IPD and NC groups. The associations between APT-related metrics and clinical assessments were investigated by Spearman correlation analysis. The receiver-operating characteristic (ROC) analysis was used to assess the diagnostic performances. The binary logistic regression model was used to combine the imaging parameters. RESULTS There wasn't any correlations between cortical APT-related signals and clinical assessment, including the H&Y scale, the disease duration, the UPDRS III scores and the MMSE scores. The MTRasym, CESTRnr and MTRRex had significantly higher values (p <0.001, corrected by Bonferroni methods) in the IPD group than NC groups in the region of bilateral and total temporal grey matter. The single parameters achieved the best diagnostic performance among all APT-related metrics was MTRRex on the right temporal grey matter, with an area under the ROC curve (AUC) of 0.865. The combined parameters achieved the highest diagnostic performance (AUC: 0.932). CONCLUSIONS 3D-APT imaging could identify the changes of the cerebral cortex in Parkinson's disease. The cortical changes of APT-related parameters could potentially serve as imaging biomarkers to aid in the non-invasive diagnosis of PD.
Collapse
Affiliation(s)
- Yaotian Tian
- Department of Radiology, National Center of Gerontology, Beijing Hospital, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730 Beijing, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, 100730 Beijing, China
| | - Xinyang Li
- Department of Radiology, National Center of Gerontology, Beijing Hospital, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730 Beijing, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, 100730 Beijing, China
| | - Xiaonan Wang
- Department of Radiology, National Center of Gerontology, Beijing Hospital, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730 Beijing, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, 100730 Beijing, China
| | - Wen Su
- Department of Neurology, National Center of Gerontology, Beijing Hospital, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730 Beijing, China
| | - Shuhua Li
- Department of Neurology, National Center of Gerontology, Beijing Hospital, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730 Beijing, China
| | - Wenqi Wang
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310027, Zhejiang, China
| | - Yi Zhang
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310027, Zhejiang, China
| | - Chunmei Li
- Department of Radiology, National Center of Gerontology, Beijing Hospital, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730 Beijing, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, 100730 Beijing, China.
| | - Min Chen
- Department of Radiology, National Center of Gerontology, Beijing Hospital, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730 Beijing, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, 100730 Beijing, China.
| |
Collapse
|
38
|
Kim H, Kim J, Sun PZ. CEST2022 - mapping multi-pool CEST signal changes in an animal model of brain tumor with quasi-steady-state reconstruction-empowered CEST quantification. Magn Reson Imaging 2023:S0730-725X(23)00100-5. [PMID: 37321379 DOI: 10.1016/j.mri.2023.06.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 06/12/2023] [Indexed: 06/17/2023]
Abstract
Chemical exchange saturated transfer (CEST) MRI has biomarker potential to assess tissue microenvironment in brain tumors. Multi-pool Lorentzian or spinlock models provides useful insights into the CEST contrast mechanism. However, T1 contribution to the complex overlapping effects of brain tumors is difficult under the non-equilibrium state. Therefore, this study evaluated T1 contributions on multi-pool parameters with quasi-steady-state (QUASS) algorithm reconstructed equilibrium data. MRI scans were performed in rat brain tumor models, including relaxation, diffusion, and CEST imaging. A pixel-wise seven-pool spinlock-model was employed to fit QUASS reconstructed CEST Z-spectra and evaluated the magnetization transfer (MT), amide, amine, guanidyl, and nuclear-overhauled effect (NOE) signals in tumor and normal tissues. In addition, T1 was estimated from the spinlock-model fitting and compared with measured T1. We observed tumor had a statistically significant increase in the amide signal (p < 0.001) and decreases in the MT and NOE signals (p < 0.001). On the other hand, the differences in amine and guanidyl between the tumor and contralateral normal regions were not statistically significant. The differences between measured and estimated T1 values were 8% in the normal tissue and 4% in the tumor. Furthermore, the isolated MT signal strongly correlated with R1 (r = 0.96, P < 0.001). In summary, we successfully unraveled multi-factorial effects in the CEST signal using spinlock-model fitting and QUASS method and demonstrated the effect of T1 relaxation on MT and NOE.
Collapse
Affiliation(s)
- Hahnsung Kim
- Emory National Primate Research Center, Emory University, Atlanta, GA, United States of America; Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States of America.
| | - Jinsuh Kim
- Emory National Primate Research Center, Emory University, Atlanta, GA, United States of America; Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Phillip Zhe Sun
- Emory National Primate Research Center, Emory University, Atlanta, GA, United States of America; Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States of America.
| |
Collapse
|
39
|
Khormi I, Al-Iedani O, Casagranda S, Papageorgakis C, Alshehri A, Lea R, Liebig P, Ramadan S, Lechner-Scott J. CEST 2022 - Differences in APT-weighted signal in T1 weighted isointense lesions, black holes and normal-appearing white matter in people with relapsing-remitting multiple sclerosis. Magn Reson Imaging 2023:S0730-725X(23)00098-X. [PMID: 37321380 DOI: 10.1016/j.mri.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 03/09/2023] [Accepted: 06/12/2023] [Indexed: 06/17/2023]
Abstract
PURPOSE To evaluate amide proton transfer weighted (APTw) signal differences between multiple sclerosis (MS) lesions and contralateral normal-appearing white matter (cNAWM). Cellular changes during the demyelination process were also assessed by comparing APTw signal intensity in T1weighted isointense (ISO) and hypointense (black hole -BH) MS lesions in relation to cNAWM. METHODS Twenty-four people with relapsing-remitting MS (pw-RRMS) on stable therapy were recruited. MRI/APTw acquisitions were undertaken on a 3 T MRI scanner. The pre and post-processing, analysis, co-registration with structural MRI maps, and identification of regions of interest (ROIs) were all performed with Olea Sphere 3.0 software. Generalized linear model (GLM) univariate ANOVA was undertaken to test the hypotheses that differences in mean APTw were entered as dependent variables. ROIs were entered as random effect variables, which allowed all data to be included. Regions (lesions and cNAWM) and/or structure (ISO and BH) were the main factor variables. The models also included age, sex, disease duration, EDSS, and ROI volumes as covariates. Receiver operating characteristic (ROC) curve analyses were performed to evaluate the diagnostic performance of these comparisons. RESULTS A total of 502 MS lesions manually identified on T2-FLAIR from twenty-four pw-RRMS were subcategorized as 359 ISO and 143 BH with reference to the T1-MPRAGE cerebral cortex signal. Also, 490 ROIs of cNAWM were manually delineated to match the MS lesion positions. A two-tailed t-test showed that mean APTw values were higher in females than in males (t = 3.52, p < 0.001). Additionally, the mean APTw values of MS lesions were higher than those of cNAWM after accounting for covariates (mean lesion = 0.44, mean cNAWM = 0.13, F = 44.12, p < 0.001).The mean APTw values of ISO lesions were higher than those of cNAWM after accounting for covariates (mean ISO lesions = 0.42, mean cNAWM = 0.21, F = 12.12, p < 0.001). The mean APTw values of BH were also higher than those of cNAWM (mean BH lesions = 0.47, mean cNAWM = 0.033, F = 40.3, p < 0.001). The effect size (i.e., difference between lesion and cNAWM) for BH was found to be higher than for ISO (14 vs. 2). Diagnostic performance showed that APT was able to discriminate between all lesions and cNAWM with an accuracy of >75% (AUC = 0.79, SE = 0.014). Discrimination between ISO lesions and cNAWM was accomplished with an accuracy of >69% (AUC = 0.74, SE = 0.018), while discrimination between BH lesions and cNAWM was achieved at an accuracy of >80% (AUC = 0.87, SE = 0.021). CONCLUSIONS Our results highlight the potential of APTw imaging for use as a non-invasive technique that is able to provide essential molecular information to clinicians and researchers so that the stages of inflammation and degeneration in MS lesions can be better characterized.
Collapse
Affiliation(s)
- Ibrahim Khormi
- School of Health Sciences, College of Health, Medicine and Wellbeing, University of Newcastle, Newcastle, Australia; Hunter Medical Research Institute, New Lambton Heights, Australia; College of Applied Medical Sciences, University of Jeddah, Jeddah, Saudi Arabia
| | - Oun Al-Iedani
- Hunter Medical Research Institute, New Lambton Heights, Australia; School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Newcastle, Australia
| | | | | | - Abdulaziz Alshehri
- School of Health Sciences, College of Health, Medicine and Wellbeing, University of Newcastle, Newcastle, Australia; Hunter Medical Research Institute, New Lambton Heights, Australia; Department of Radiology, King Fahd Hospital of the University, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Rodney Lea
- Hunter Medical Research Institute, New Lambton Heights, Australia
| | | | - Saadallah Ramadan
- School of Health Sciences, College of Health, Medicine and Wellbeing, University of Newcastle, Newcastle, Australia; Hunter Medical Research Institute, New Lambton Heights, Australia.
| | - Jeannette Lechner-Scott
- Hunter Medical Research Institute, New Lambton Heights, Australia; Department of Neurology, John Hunter Hospital, New Lambton Heights, Australia; School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Newcastle, Australia
| |
Collapse
|
40
|
Igarashi T, Kim H, Sun PZ. Detection of tissue pH with quantitative chemical exchange saturation transfer magnetic resonance imaging. NMR IN BIOMEDICINE 2023; 36:e4711. [PMID: 35141979 PMCID: PMC10249910 DOI: 10.1002/nbm.4711] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 02/03/2022] [Accepted: 02/05/2022] [Indexed: 05/12/2023]
Abstract
Chemical exchange saturation transfer (CEST) magnetic resonance imaging (MRI) has emerged as a novel means for sensitive detection of dilute labile protons and chemical exchange rates. By sensitizing to pH-dependent chemical exchange, CEST MRI has shown promising results in monitoring tissue statuses such as pH changes in disorders like acute stroke, tumor, and acute kidney injury. This article briefly reviews the basic principles for CEST imaging and quantitative measures, from the simplistic asymmetry analysis to multipool Lorentzian decoupling and quasi-steady-state reconstruction. In particular, the advantages and limitations of commonly used quantitative approaches for CEST applications are discussed.
Collapse
Affiliation(s)
- Takahiro Igarashi
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA
| | - Hahnsung Kim
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
- Yerkes Imaging Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Phillip Zhe Sun
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
- Yerkes Imaging Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| |
Collapse
|
41
|
Perlman O, Farrar CT, Heo HY. MR fingerprinting for semisolid magnetization transfer and chemical exchange saturation transfer quantification. NMR IN BIOMEDICINE 2023; 36:e4710. [PMID: 35141967 PMCID: PMC9808671 DOI: 10.1002/nbm.4710] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 01/18/2022] [Accepted: 02/04/2022] [Indexed: 05/11/2023]
Abstract
Chemical exchange saturation transfer (CEST) MRI has positioned itself as a promising contrast mechanism, capable of providing molecular information at sufficient resolution and amplified sensitivity. However, it has not yet become a routinely employed clinical technique, due to a variety of confounding factors affecting its contrast-weighted image interpretation and the inherently long scan time. CEST MR fingerprinting (MRF) is a novel approach for addressing these challenges, allowing simultaneous quantitation of several proton exchange parameters using rapid acquisition schemes. Recently, a number of deep-learning algorithms have been developed to further boost the performance and speed of CEST and semi-solid macromolecule magnetization transfer (MT) MRF. This review article describes the fundamental theory behind semisolid MT/CEST-MRF and its main applications. It then details supervised and unsupervised learning approaches for MRF image reconstruction and describes artificial intelligence (AI)-based pipelines for protocol optimization. Finally, practical considerations are discussed, and future perspectives are given, accompanied by basic demonstration code and data.
Collapse
Affiliation(s)
- Or Perlman
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Christian T. Farrar
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Hye-Young Heo
- Division of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| |
Collapse
|
42
|
Jiang S, Wen Z, Ahn SS, Cai K, Paech D, Eberhart CG, Zhou J. Applications of chemical exchange saturation transfer magnetic resonance imaging in identifying genetic markers in gliomas. NMR IN BIOMEDICINE 2023; 36:e4731. [PMID: 35297117 PMCID: PMC10557022 DOI: 10.1002/nbm.4731] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/07/2022] [Accepted: 03/14/2022] [Indexed: 05/23/2023]
Abstract
Chemical exchange saturation transfer (CEST) imaging is an important molecular magnetic resonance imaging technique that can image numerous low-concentration biomolecules with water-exchangeable protons (such as cellular proteins) and tissue pH. CEST, or more specially amide proton transfer-weighted imaging, has been widely used for the detection, diagnosis, and response assessment of brain tumors, and its feasibility in identifying molecular markers in gliomas has also been explored in recent years. In this paper, after briefing on the basic principles and quantification methods of CEST imaging, we review its early applications in identifying isocitrate dehydrogenase mutation status, MGMT methylation status, 1p/19q deletion status, and H3K27M mutation status in gliomas. Finally, we discuss the limitations or weaknesses in these studies.
Collapse
Affiliation(s)
- Shanshan Jiang
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
| | - Zhibo Wen
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Sung Soo Ahn
- Department of Radiology and Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Kejia Cai
- Department of Radiology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Daniel Paech
- Department of Radiology, German Cancer Research Center, Heidelberg, Germany
- Clinic for Neuroradiology, University Hospital Bonn, Bonn, Germany
| | | | - Jinyuan Zhou
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
43
|
Cember ATJ, Nanga RPR, Reddy R. Glutamate-weighted CEST (gluCEST) imaging for mapping neurometabolism: An update on the state of the art and emerging findings from in vivo applications. NMR IN BIOMEDICINE 2023; 36:e4780. [PMID: 35642353 DOI: 10.1002/nbm.4780] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 05/23/2023]
Abstract
Glutamate is the primary excitatory neurotransmitter in the mammalian central nervous system. As such, its proper regulation is essential to the healthy function of the human brain, and dysregulation of glutamate metabolism and compartmentalization underlies numerous neurological and neuropsychiatric pathologies. Glutamate-weighted chemical exchange saturation transfer (gluCEST) MRI is one of the only ways to non-invasively observe the relative concentration and spatial distribution of glutamate in the human brain. In the past 10 years, gluCEST has developed from a proof-of-concept experiment carried out in imaging phantoms and model systems to an increasingly sophisticated technique applied to reveal deviations from baseline neural metabolism in human beings, most notably in patients experiencing seizures of various origins or those on the psychosis spectrum. This article traces that progress, including in-depth discussion of the technical specifics of gluCEST and potential challenges to performing these experiments rigorously. We discuss the neurobiological context of glutamate, including the widely accepted hypotheses and models in the literature regarding its involvement in neurodegenerative diseases and other pathology. We then review the state of the art of in vivo glutamate detection by magnetic resonance imaging and the limitations on this front of in vivo MR spectroscopy. The gluCEST experiment is introduced and its advantages, challenges and limitations are thoroughly explored, beginning with the phantom experiment results demonstrated in the initial publication, through the latest approaches to correcting human brain images for B1 inhomogeneity. We then give a comprehensive overview of preclinical applications demonstrated to date, including Alzheimer's disease, Parkinson's disease, Huntington's disease, Traumatic brain injury and cancer, followed by a similar discussion of human studies. Finally, we highlight emerging applications, and discuss technical improvements on the horizon that hold promise for improving the robustness and versatility of gluCEST and its increasing presence in the arena of translational and precision medicine.
Collapse
Affiliation(s)
- Abigail T J Cember
- Center for Advanced Metabolic Imaging in Precision Medicine (CAMIPM), Department of Radiology, University of Pennsylvania
| | - Ravi Prakash Reddy Nanga
- Center for Advanced Metabolic Imaging in Precision Medicine (CAMIPM), Department of Radiology, University of Pennsylvania
| | - Ravinder Reddy
- Center for Advanced Metabolic Imaging in Precision Medicine (CAMIPM), Department of Radiology, University of Pennsylvania
| |
Collapse
|
44
|
Hangel G, Schmitz‐Abecassis B, Sollmann N, Pinto J, Arzanforoosh F, Barkhof F, Booth T, Calvo‐Imirizaldu M, Cassia G, Chmelik M, Clement P, Ercan E, Fernández‐Seara MA, Furtner J, Fuster‐Garcia E, Grech‐Sollars M, Guven NT, Hatay GH, Karami G, Keil VC, Kim M, Koekkoek JAF, Kukran S, Mancini L, Nechifor RE, Özcan A, Ozturk‐Isik E, Piskin S, Schmainda KM, Svensson SF, Tseng C, Unnikrishnan S, Vos F, Warnert E, Zhao MY, Jancalek R, Nunes T, Hirschler L, Smits M, Petr J, Emblem KE. Advanced MR Techniques for Preoperative Glioma Characterization: Part 2. J Magn Reson Imaging 2023; 57:1676-1695. [PMID: 36912262 PMCID: PMC10947037 DOI: 10.1002/jmri.28663] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 03/14/2023] Open
Abstract
Preoperative clinical MRI protocols for gliomas, brain tumors with dismal outcomes due to their infiltrative properties, still rely on conventional structural MRI, which does not deliver information on tumor genotype and is limited in the delineation of diffuse gliomas. The GliMR COST action wants to raise awareness about the state of the art of advanced MRI techniques in gliomas and their possible clinical translation. This review describes current methods, limits, and applications of advanced MRI for the preoperative assessment of glioma, summarizing the level of clinical validation of different techniques. In this second part, we review magnetic resonance spectroscopy (MRS), chemical exchange saturation transfer (CEST), susceptibility-weighted imaging (SWI), MRI-PET, MR elastography (MRE), and MR-based radiomics applications. The first part of this review addresses dynamic susceptibility contrast (DSC) and dynamic contrast-enhanced (DCE) MRI, arterial spin labeling (ASL), diffusion-weighted MRI, vessel imaging, and magnetic resonance fingerprinting (MRF). EVIDENCE LEVEL: 3. TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Gilbert Hangel
- Department of NeurosurgeryMedical University of ViennaViennaAustria
- High Field MR Centre, Department of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
- Christian Doppler Laboratory for MR Imaging BiomarkersViennaAustria
- Medical Imaging ClusterMedical University of ViennaViennaAustria
| | - Bárbara Schmitz‐Abecassis
- Department of RadiologyLeiden University Medical CenterLeidenthe Netherlands
- Medical Delta FoundationDelftthe Netherlands
| | - Nico Sollmann
- Department of Diagnostic and Interventional RadiologyUniversity Hospital UlmUlmGermany
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der IsarTechnical University of MunichMunichGermany
- TUM‐Neuroimaging Center, Klinikum rechts der IsarTechnical University of MunichMunichGermany
| | - Joana Pinto
- Institute of Biomedical Engineering, Department of Engineering ScienceUniversity of OxfordOxfordUK
| | | | - Frederik Barkhof
- Department of Radiology & Nuclear MedicineAmsterdam UMC, Vrije UniversiteitAmsterdamNetherlands
- Queen Square Institute of Neurology and Centre for Medical Image ComputingUniversity College LondonLondonUK
| | - Thomas Booth
- School of Biomedical Engineering and Imaging SciencesKing's College LondonLondonUK
- Department of NeuroradiologyKing's College Hospital NHS Foundation TrustLondonUK
| | | | | | - Marek Chmelik
- Department of Technical Disciplines in Medicine, Faculty of Health CareUniversity of PrešovPrešovSlovakia
| | - Patricia Clement
- Department of Diagnostic SciencesGhent UniversityGhentBelgium
- Department of Medical ImagingGhent University HospitalGhentBelgium
| | - Ece Ercan
- Department of RadiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Maria A. Fernández‐Seara
- Department of RadiologyClínica Universidad de NavarraPamplonaSpain
- IdiSNA, Instituto de Investigación Sanitaria de NavarraPamplonaSpain
| | - Julia Furtner
- Department of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
- Research Center of Medical Image Analysis and Artificial IntelligenceDanube Private UniversityAustria
| | - Elies Fuster‐Garcia
- Biomedical Data Science Laboratory, Instituto Universitario de Tecnologías de la Información y ComunicacionesUniversitat Politècnica de ValènciaValenciaSpain
| | - Matthew Grech‐Sollars
- Centre for Medical Image Computing, Department of Computer ScienceUniversity College LondonLondonUK
- Lysholm Department of Neuroradiology, National Hospital for Neurology and NeurosurgeryUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - N. Tugay Guven
- Institute of Biomedical EngineeringBogazici University IstanbulIstanbulTurkey
| | - Gokce Hale Hatay
- Institute of Biomedical EngineeringBogazici University IstanbulIstanbulTurkey
| | - Golestan Karami
- School of Biomedical Engineering and Imaging SciencesKing's College LondonLondonUK
| | - Vera C. Keil
- Department of Radiology & Nuclear MedicineAmsterdam UMC, Vrije UniversiteitAmsterdamNetherlands
- Cancer Center AmsterdamAmsterdamNetherlands
| | - Mina Kim
- Centre for Medical Image Computing, Department of Medical Physics & Biomedical Engineering and Department of NeuroinflammationUniversity College LondonLondonUK
| | - Johan A. F. Koekkoek
- Department of NeurologyLeiden University Medical CenterLeidenthe Netherlands
- Department of NeurologyHaaglanden Medical CenterNetherlands
| | - Simran Kukran
- Department of BioengineeringImperial College LondonLondonUK
- Department of Radiotherapy and ImagingInstitute of Cancer ResearchUK
| | - Laura Mancini
- Lysholm Department of Neuroradiology, National Hospital for Neurology and NeurosurgeryUniversity College London Hospitals NHS Foundation TrustLondonUK
- Department of Brain Repair and Rehabilitation, Institute of NeurologyUniversity College LondonLondonUK
| | - Ruben Emanuel Nechifor
- Department of Clinical Psychology and Psychotherapy, International Institute for the Advanced Studies of Psychotherapy and Applied Mental HealthBabes‐Bolyai UniversityRomania
| | - Alpay Özcan
- Electrical and Electronics Engineering DepartmentBogazici University IstanbulIstanbulTurkey
| | - Esin Ozturk‐Isik
- Institute of Biomedical EngineeringBogazici University IstanbulIstanbulTurkey
| | - Senol Piskin
- Department of Mechanical Engineering, Faculty of Natural Sciences and EngineeringIstinye University IstanbulIstanbulTurkey
| | | | - Siri F. Svensson
- Department of Physics and Computational RadiologyOslo University HospitalOsloNorway
- Department of PhysicsUniversity of OsloOsloNorway
| | - Chih‐Hsien Tseng
- Medical Delta FoundationDelftthe Netherlands
- Department of Imaging PhysicsDelft University of TechnologyDelftthe Netherlands
| | - Saritha Unnikrishnan
- Faculty of Engineering and DesignAtlantic Technological University (ATU) SligoSligoIreland
- Mathematical Modelling and Intelligent Systems for Health and Environment (MISHE), ATU SligoSligoIreland
| | - Frans Vos
- Medical Delta FoundationDelftthe Netherlands
- Department of Radiology & Nuclear MedicineErasmus MCRotterdamNetherlands
- Department of Imaging PhysicsDelft University of TechnologyDelftthe Netherlands
| | - Esther Warnert
- Department of Radiology & Nuclear MedicineErasmus MCRotterdamNetherlands
| | - Moss Y. Zhao
- Department of RadiologyStanford UniversityStanfordCaliforniaUSA
- Stanford Cardiovascular InstituteStanford UniversityStanfordCaliforniaUSA
| | - Radim Jancalek
- Department of NeurosurgerySt. Anne's University HospitalBrnoCzechia
- Faculty of MedicineMasaryk UniversityBrnoCzechia
| | - Teresa Nunes
- Department of NeuroradiologyHospital Garcia de OrtaAlmadaPortugal
| | - Lydiane Hirschler
- C.J. Gorter MRI Center, Department of RadiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Marion Smits
- Medical Delta FoundationDelftthe Netherlands
- Department of Radiology & Nuclear MedicineErasmus MCRotterdamNetherlands
- Brain Tumour CentreErasmus MC Cancer InstituteRotterdamthe Netherlands
| | - Jan Petr
- Helmholtz‐Zentrum Dresden‐RossendorfInstitute of Radiopharmaceutical Cancer ResearchDresdenGermany
| | - Kyrre E. Emblem
- Department of Physics and Computational RadiologyOslo University HospitalOsloNorway
| |
Collapse
|
45
|
Heo HY, Tee YK, Harston G, Leigh R, Chappell M. Amide proton transfer imaging in stroke. NMR IN BIOMEDICINE 2023; 36:e4734. [PMID: 35322482 PMCID: PMC9761584 DOI: 10.1002/nbm.4734] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/04/2022] [Accepted: 03/21/2022] [Indexed: 05/23/2023]
Abstract
Amide proton transfer (APT) imaging, a variant of chemical exchange saturation transfer MRI, has shown promise in detecting ischemic tissue acidosis following impaired aerobic metabolism in animal models and in human stroke patients due to the sensitivity of the amide proton exchange rate to changes in pH within the physiological range. Recent studies have demonstrated the possibility of using APT-MRI to detect acidosis of the ischemic penumbra, enabling the assessment of stroke severity and risk of progression, monitoring of treatment progress, and prognostication of clinical outcome. This paper reviews current APT imaging methods actively used in ischemic stroke research and explores the clinical aspects of ischemic stroke and future applications for these methods.
Collapse
Affiliation(s)
- Hye-Young Heo
- Division of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Yee Kai Tee
- Lee Kong Chian Faculty of Engineering and Science, University Tunku Abdul Rahman, Malaysia
| | - George Harston
- Acute Stroke Programme, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Richard Leigh
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael Chappell
- Radiological Sciences, Mental Health and Clinical Neurosciences, School of Medicine, University of Nottingham, Nottingham, UK
- Nottingham Biomedical Research Centre, Queen’s Medical Centre, University of Nottingham, Nottingham, United Kingdom, UK
| |
Collapse
|
46
|
Zhou Y, Bie C, van Zijl PC, Yadav NN. The relayed nuclear Overhauser effect in magnetization transfer and chemical exchange saturation transfer MRI. NMR IN BIOMEDICINE 2023; 36:e4778. [PMID: 35642102 PMCID: PMC9708952 DOI: 10.1002/nbm.4778] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/19/2022] [Accepted: 05/29/2022] [Indexed: 05/23/2023]
Abstract
Magnetic resonance (MR) is a powerful technique for noninvasively probing molecular species in vivo but suffers from low signal sensitivity. Saturation transfer (ST) MRI approaches, including chemical exchange saturation transfer (CEST) and conventional magnetization transfer contrast (MTC), allow imaging of low-concentration molecular components with enhanced sensitivity using indirect detection via the abundant water proton pool. Several recent studies have shown the utility of chemical exchange relayed nuclear Overhauser effect (rNOE) contrast originating from nonexchangeable carbon-bound protons in mobile macromolecules in solution. In this review, we describe the mechanisms leading to the occurrence of rNOE-based signals in the water saturation spectrum (Z-spectrum), including those from mobile and immobile molecular sources and from molecular binding. While it is becoming clear that MTC is mainly an rNOE-based signal, we continue to use the classical MTC nomenclature to separate it from the rNOE signals of mobile macromolecules, which we will refer to as rNOEs. Some emerging applications of the use of rNOEs for probing macromolecular solution components such as proteins and carbohydrates in vivo or studying the binding of small substrates are discussed.
Collapse
Affiliation(s)
- Yang Zhou
- Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, Guangdong 518055 (China)
| | - Chongxue Bie
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, 707 N. Broadway, Baltimore MD 21205 (USA)
- The Russell H. Morgan Department of Radiology, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Baltimore, MD 21205 (USA)
- Department of Information Science and Technology, Northwest University, No.1 Xuefu Avenue, Xi’an, Shanxi 710127 (China)
| | - Peter C.M. van Zijl
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, 707 N. Broadway, Baltimore MD 21205 (USA)
- The Russell H. Morgan Department of Radiology, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Baltimore, MD 21205 (USA)
| | - Nirbhay N. Yadav
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, 707 N. Broadway, Baltimore MD 21205 (USA)
- The Russell H. Morgan Department of Radiology, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Baltimore, MD 21205 (USA)
| |
Collapse
|
47
|
Liu Y, Gauthier GC, Gendelman HE, Bade AN. Dual-Peak Lorentzian CEST MRI for antiretroviral drug brain distribution. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2023; 2:63-69. [PMID: 37027345 PMCID: PMC10070014 DOI: 10.1515/nipt-2022-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 08/24/2022] [Indexed: 11/15/2022]
Abstract
Objectives Spatial-temporal biodistribution of antiretroviral drugs (ARVs) can now be achieved using MRI by utilizing chemical exchange saturation transfer (CEST) contrasts. However, the presence of biomolecules in tissue limits the specificity of current CEST methods. To overcome this limitation, a Lorentzian line-shape fitting algorithm was developed that simultaneously fits CEST peaks of ARV protons on its Z-spectrum. Case presentation This algorithm was tested on the common first line ARV, lamivudine (3TC), that has two peaks resulting from amino (-NH2) and hydroxyl (-OH) protons in 3TC. The developed dual-peak Lorentzian function fitted these two peaks simultaneously, and used the ratio of -NH2 and -OH CEST contrasts as a constraint parameter to measure 3TC presence in brains of drug-treated mice. 3TC biodistribution calculated using the new algorithm was compared against actual drug levels measured using UPLC-MS/MS. In comparison to the method that employs the -NH2 CEST peak only, the dual-peak Lorentzian fitting algorithm showed stronger correlation with brain tissue 3TC levels, signifying estimation of actual drug levels. Conclusions We concluded that 3TC levels can be extracted from confounding CEST effects of tissue biomolecules resulting in improved specificity for drug mapping. This algorithm can be expanded to measure a variety of ARVs using CEST MRI.
Collapse
Affiliation(s)
- Yutong Liu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Radiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Gabriel C. Gauthier
- Department of Radiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Aditya N. Bade
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
48
|
Sun PZ. Demonstration of accurate multi-pool chemical exchange saturation transfer MRI quantification - Quasi-steady-state reconstruction empowered quantitative CEST analysis. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2023; 348:107379. [PMID: 36689786 PMCID: PMC10023465 DOI: 10.1016/j.jmr.2023.107379] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/05/2023] [Accepted: 01/15/2023] [Indexed: 05/18/2023]
Abstract
Chemical exchange saturation transfer (CEST) MRI is sensitive to dilute labile protons and microenvironment properties, yet CEST quantification has been challenging. This difficulty is because the CEST measurement depends not only on the underlying CEST system but also on the scan protocols, including RF saturation amplitude, duration, and repetition time. In addition, T1 normalization is not straightforward under non-equilibrium conditions. Recently, a quasi-steady-state (QUASS) algorithm was established to reconstruct the desired equilibrium state from experimental measurements. Our study aimed to determine the accuracy of spinlock-model-based multi-pool CEST quantification using numerical simulations and phantom experiments. In short, CEST Z-spectra were simulated for a representative 3-pool model, and CEST amplitudes were solved with spinlock model-based multi-pool fitting and assessed as a function of RF saturation time (Ts), repetition time (TR), and T1. Although the apparent CEST signals showed significant T1 dependence, such relationships were not observed following QUASS reconstruction. To test the accuracy of T1 correction, a multi-vial phantom of nicotinamide and creatine was doped with manganese chloride, resulting in T1 ranging from 1 s to beyond 2 s. The multi-labile signals determined from the routine measurements showed significant dependence on Ts, TR, and T1. In contrast, CEST signals from the QUASS reconstruction showed consistent quantification independent of such variables. To summarize, our study demonstrated that accurate CEST quantification is feasible in multi-pool CEST systems with the spinlock-model-based fitting of QUASS CEST MRI.
Collapse
Affiliation(s)
- Phillip Zhe Sun
- Primate Imaging Center, Emory National Primate Research Center, Emory University, Atlanta, GA, United States; Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States.
| |
Collapse
|
49
|
Zu T, Sun Y, Wu D, Zhang Y. Joint K-space and Image-space Parallel Imaging (KIPI) for accelerated chemical exchange saturation transfer acquisition. Magn Reson Med 2023; 89:922-936. [PMID: 36336741 DOI: 10.1002/mrm.29480] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/25/2022] [Accepted: 09/16/2022] [Indexed: 11/09/2022]
Abstract
PURPOSE To develop an auto-calibrated technique by joint K-space and Image-space Parallel Imaging (KIPI) for accelerated CEST acquisition. THEORY AND METHODS The KIPI method selects a calibration frame with a low acceleration factor (AF) and auto-calibration signals (ACS) acquired, from which the coil sensitivity profiles and artifact correction maps are calculated after restoring the k-space by GRAPPA. Then the other frames with high AF and without ACS can be reconstructed by SENSE and artifact suppression. The signal leakage due to the T2 -decay filtering in k-space compromises the SENSE reconstruction, which can be corrected by the artifact suppression algorithm of KIPI. The 2D and 3D imaging experiments were done on the phantom, healthy volunteer, and brain tumor patient with a 3T scanner. RESULTS The proposed KIPI method was evaluated by retrospectively undersampled data with variable AFs and compared against existing parallel imaging methods (SENSE/auto, GRAPPA, and ESPIRiT). KIPI enabled CEST frames with random AFs to achieve similar image quality, eliminated the strong aliasing artifacts, and generated significantly smaller errors than the other methods (p < 0.01). The KIPI method permitted an AF up to 12-fold in both phase-encoding and slice-encoding directions for 3D CEST source images, achieving an overall 8.2-fold speedup in scan time. CONCLUSION KIPI is a novel auto-calibrated parallel imaging method that enables variable AFs for different CEST frames, achieves a significant reduction in scan time, and does not compromise the accuracy of CEST maps.
Collapse
Affiliation(s)
- Tao Zu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yi Sun
- MR Collaboration, Siemens Healthcare Ltd., Shanghai, China
| | - Dan Wu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yi Zhang
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
50
|
Moore-Palhares D, Ho L, Lu L, Chugh B, Vesprini D, Karam I, Soliman H, Symons S, Leung E, Loblaw A, Myrehaug S, Stanisz G, Sahgal A, Czarnota GJ. Clinical implementation of magnetic resonance imaging simulation for radiation oncology planning: 5 year experience. Radiat Oncol 2023; 18:27. [PMID: 36750891 PMCID: PMC9903411 DOI: 10.1186/s13014-023-02209-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 01/16/2023] [Indexed: 02/09/2023] Open
Abstract
PURPOSE Integrating magnetic resonance (MR) into radiotherapy planning has several advantages. This report details the clinical implementation of an MR simulation (MR-planning) program for external beam radiotherapy (EBRT) in one of North America's largest radiotherapy programs. METHODS AND MATERIALS An MR radiotherapy planning program was developed and implemented at Sunnybrook Health Sciences Center in 2016 with two dedicated wide-bore MR platforms (1.5 and 3.0 Tesla). Planning MR was sequentially implemented every 3 months for separate treatment sites, including the central nervous system (CNS), gynecologic (GYN), head and neck (HN), genitourinary (GU), gastrointestinal (GI), breast, and brachial plexus. Essential protocols and processes were detailed in this report, including clinical workflow, optimized MR-image acquisition protocols, MR-adapted patient setup, strategies to overcome risks and challenges, and an MR-planning quality assurance program. This study retrospectively reviewed simulation site data for all MR-planning sessions performed for EBRT over the past 5 years. RESULTS From July 2016 to December 2021, 8798 MR-planning sessions were carried out, which corresponds to 25% of all computer tomography (CT) simulations (CT-planning) performed during the same period at our institution. There was a progressive rise from 80 MR-planning sessions in 2016 to 1126 in 2017, 1492 in 2018, 1824 in 2019, 2040 in 2020, and 2236 in 2021. As a result, the relative number of planning MR/CT increased from 3% of all planning sessions in 2016 to 36% in 2021. The most common site of MR-planning was CNS (49%), HN (13%), GYN (12%), GU (12%), and others (8%). CONCLUSION Detailed clinical processes and protocols of our MR-planning program were presented, which have been improved over more than 5 years of robust experience. Strategies to overcome risks and challenges in the implementation process are highlighted. Our work provides details that can be used by institutions interested in implementing an MR-planning program.
Collapse
Affiliation(s)
- Daniel Moore-Palhares
- grid.413104.30000 0000 9743 1587Department of Radiation Oncology, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, T2, Toronto, ON M4N3M5 Canada ,grid.17063.330000 0001 2157 2938Department of Radiation Oncology, University of Toronto, Toronto, Canada ,grid.17063.330000 0001 2157 2938Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Ling Ho
- grid.413104.30000 0000 9743 1587Department of Radiation Oncology, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, T2, Toronto, ON M4N3M5 Canada
| | - Lin Lu
- grid.413104.30000 0000 9743 1587Department of Radiation Oncology, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, T2, Toronto, ON M4N3M5 Canada
| | - Brige Chugh
- grid.413104.30000 0000 9743 1587Department of Radiation Oncology, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, T2, Toronto, ON M4N3M5 Canada ,grid.17063.330000 0001 2157 2938Department of Radiation Oncology, University of Toronto, Toronto, Canada ,grid.17063.330000 0001 2157 2938Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Danny Vesprini
- grid.413104.30000 0000 9743 1587Department of Radiation Oncology, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, T2, Toronto, ON M4N3M5 Canada ,grid.17063.330000 0001 2157 2938Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Irene Karam
- grid.413104.30000 0000 9743 1587Department of Radiation Oncology, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, T2, Toronto, ON M4N3M5 Canada ,grid.17063.330000 0001 2157 2938Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Hany Soliman
- grid.413104.30000 0000 9743 1587Department of Radiation Oncology, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, T2, Toronto, ON M4N3M5 Canada ,grid.17063.330000 0001 2157 2938Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Sean Symons
- grid.17063.330000 0001 2157 2938Physical Sciences, Sunnybrook Research Institute, Toronto, Canada ,grid.413104.30000 0000 9743 1587Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Eric Leung
- grid.413104.30000 0000 9743 1587Department of Radiation Oncology, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, T2, Toronto, ON M4N3M5 Canada ,grid.17063.330000 0001 2157 2938Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Andrew Loblaw
- grid.413104.30000 0000 9743 1587Department of Radiation Oncology, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, T2, Toronto, ON M4N3M5 Canada ,grid.17063.330000 0001 2157 2938Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Sten Myrehaug
- grid.413104.30000 0000 9743 1587Department of Radiation Oncology, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, T2, Toronto, ON M4N3M5 Canada ,grid.17063.330000 0001 2157 2938Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Greg Stanisz
- grid.17063.330000 0001 2157 2938Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Arjun Sahgal
- grid.413104.30000 0000 9743 1587Department of Radiation Oncology, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, T2, Toronto, ON M4N3M5 Canada ,grid.17063.330000 0001 2157 2938Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Gregory J. Czarnota
- grid.413104.30000 0000 9743 1587Department of Radiation Oncology, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, T2, Toronto, ON M4N3M5 Canada ,grid.17063.330000 0001 2157 2938Department of Radiation Oncology, University of Toronto, Toronto, Canada ,grid.17063.330000 0001 2157 2938Physical Sciences, Sunnybrook Research Institute, Toronto, Canada ,grid.17063.330000 0001 2157 2938Department of Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|