1
|
Li Y, Ta L, Wu Q, Zhang H, Xu Y, Gan L, Liu J. Assessment of the Validity of Carbon Ion Irradiation for C6 Gliomas in Rats. Dose Response 2025; 23:15593258251327505. [PMID: 40297665 PMCID: PMC12033543 DOI: 10.1177/15593258251327505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 01/15/2025] [Accepted: 02/18/2025] [Indexed: 04/30/2025] Open
Abstract
Purpose Application of energy-spectrum computed tomography (CT) to assess specific efficacy of and response to carbon ion radiotherapy (CIRT) of C6 gliomas in rats. Methods After establishing C6 glioma rat models, 3 tumor-bearing rats were randomly selected as controls. The remaining were divided into 0 Gy, 1 Gy, and 2 Gy groups for CIRT. Energy-spectrum CT scans were performed, and brain tissues were collected for histopathology and western blot Test. Survival rates in each group were compared. Results The results demonstrated that tumors in the 1 Gy and 2 Gy groups decreased at different rates up to 14 days post-CIRT (P < 0.05). Furthermore, compared to pre-CIRT measurements, the energy-spectrum parameters gradually increased in the 0 Gy group, while they decreased in the 2 Gy group. Post-CIRT, the Ki-67 proliferation index and the expression levels of vascu-larassociated proteins in tumor tissues were significantly reduced in the 1 and 2 Gy groups. Additionally, the survival times of tumor-bearing rats were prolonged after CIRT. Conclusions CIRT effectively restricts tumor cell growth and proliferation, leading to improved survival rates in rats with C6 gliomas. The use of energy-spectrum CT with immunohistochemistry for quantitative detection can actively support the effectiveness of carbon ion radiotherapy in inhibiting tumor proliferation.
Collapse
Affiliation(s)
- Yufeng Li
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Lei Ta
- Ningbo Medical Center LiHuiLi Hospital, Ningbo, China
| | - QingFeng Wu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China
| | - Hongyu Zhang
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Yuan Xu
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Lu Gan
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China
| | - Jianli Liu
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
2
|
Nichelli L, Casagranda S, Dipasquale O, Bensemain M, Papageorgakis C, Zucchelli M, Jacob J, Valery C, Mathon B, Liebig P, Zaiss M, Lehéricy S. Fluid-Suppressed Amide Proton Transfer-Weighted Imaging Outperforms Leakage-Corrected Dynamic Susceptibility Contrast Perfusion in Distinguishing Progression from Radionecrosis in Brain Metastases. Cancers (Basel) 2025; 17:1175. [PMID: 40227736 PMCID: PMC11987914 DOI: 10.3390/cancers17071175] [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: 02/17/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/15/2025] Open
Abstract
Background: Differentiating brain radionecrosis (RN) from tumor progression (TP) is a persistent clinical difficulty. Here, we compared the diagnostic accuracy of leakage-corrected relative cerebral blood volume (rCBV) and fluid-suppressed amide proton transfer-weighted (APTw) imaging in distinguishing between RN and TP in metastases. Methods: Subjects with enlarging lesions after stereotactic radiosurgery were prospectively examined at 3T. APTw data were acquired with a 3D snapshot-gradient echo sequence. B0 and B1 inhomogeneities were corrected using the WASAB1 protocol. rCBV was calculated according to established guidelines. Image analysis was performed using Olea Sphere 3.0 software. ΔAPTw and ΔrCBV were calculated as the average signal within the lesion normalized against the average signal in the contralateral white matter. A diagnosis of TP or RN was assessed by histology or imaging at follow-up. Independent samples t-tests of ΔAPTw and ΔrCBV and the areas under the curve (AUCs) were computed. Results: Twenty-one metastases (10 RN, 11 TP) were evaluated. APTw differentiated between RN and TP (U = 120, p < 0.001), in contrast to rCBV (U = 71, p = 0.174). The AUC was 0.991 (95% CI = 0.962-1.020) for ΔAPTw, and 0.636 (95% CI = 0.352-0.921) for ΔrCBV. The optimal cutoff points were 0.4 and 2.1 for ΔAPTw and ΔrCBV, respectively. The sensitivity and specificity for RN-TP were 100% and 90% for ΔAPTw and 63.6% and 36.4% for ΔrCBV. Conclusions: Fluid-suppressed APTw metrics enabled more accurate diagnostic performances than leakage-corrected rCBV metrics in distinguishing between RN and TP. These promising results suggest that APTw imaging could valuably complement current multiparametric MRI protocols in brain metastases follow-ups.
Collapse
Affiliation(s)
- Lucia Nichelli
- Paris Brain Institute (PBI)—Institut du Cerveau (ICM), Sorbonne University, Inserm UMR S 1127, CNRS 7225, F-75013 Paris, France
- Department of Neuroradiology, La Pitié Salpêtrière Hospital, AP-HP Sorbonne University, F-75013 Paris, France
| | - Stefano Casagranda
- Department of R&D Advanced Applications, Olea Medical, 13600 La Ciotat, France
| | - Ottavia Dipasquale
- Department of R&D Advanced Applications, Olea Medical, 13600 La Ciotat, France
| | - Mehdi Bensemain
- Department of Neuroradiology, La Pitié Salpêtrière Hospital, AP-HP Sorbonne University, F-75013 Paris, France
| | | | - Mauro Zucchelli
- Department of R&D Advanced Applications, Olea Medical, 13600 La Ciotat, France
| | - Julian Jacob
- Department of Radiation-Oncology, La Pitié Salpêtrière Hospital, AP-HP Sorbonne University, F-75013 Paris, France
| | - Charles Valery
- Department of Neurosurgery, La Pitié Salpêtrière Hospital, AP-HP Sorbonne University, F-75013 Paris, France
| | - Bertrand Mathon
- Paris Brain Institute (PBI)—Institut du Cerveau (ICM), Sorbonne University, Inserm UMR S 1127, CNRS 7225, F-75013 Paris, France
- Department of Neurosurgery, La Pitié Salpêtrière Hospital, AP-HP Sorbonne University, F-75013 Paris, France
| | | | - Moritz Zaiss
- Department of Neuroradiology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Stéphane Lehéricy
- Paris Brain Institute (PBI)—Institut du Cerveau (ICM), Sorbonne University, Inserm UMR S 1127, CNRS 7225, F-75013 Paris, France
- Department of Neuroradiology, La Pitié Salpêtrière Hospital, AP-HP Sorbonne University, F-75013 Paris, France
| |
Collapse
|
3
|
Laudicella R, Mantarro C, Catalfamo B, Alongi P, Gaeta M, Minutoli F, Baldari S, Bisdas S. PET Imaging in Gliomas. RADIOLOGY‐NUCLEAR MEDICINE DIAGNOSTIC IMAGING 2023:194-218. [DOI: 10.1002/9781119603627.ch6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Koike H, Morikawa M, Ishimaru H, Ideguchi R, Uetani M, Hiu T, Matsuo T, Miyoshi M. Amide proton transfer MRI differentiates between progressive multifocal leukoencephalopathy and malignant brain tumors: a pilot study. BMC Med Imaging 2022; 22:227. [PMID: 36572873 PMCID: PMC9793649 DOI: 10.1186/s12880-022-00959-3] [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: 09/08/2022] [Accepted: 12/22/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Progressive multifocal leukoencephalopathy (PML) is a demyelinating disease of the central nerve system caused by the John Cunningham virus. On MRI, PML may sometimes appear similar to primary central nervous system lymphoma (PCNSL) and glioblastoma multiforme (GBM). The purpose of this pilot study was to evaluate the potential of amide proton transfer (APT) imaging for differentiating PML from PCNSL and GBM. METHODS Patients with PML (n = 4; two men; mean age 52.3 ± 6.1 years), PCNSL (n = 7; four women; mean age 74.4 ± 5.8 years), or GBM (n = 11; 6 men; mean age 65.0 ± 15.2 years) who underwent APT-CEST MRI between January 2021 and September 2022 were retrospectively evaluated. Magnetization transfer ratio asymmetry (MTRasym) values were measured on APT imaging using a region of interest within the lesion. Receiver operating characteristics curve analysis was used to determine diagnostic cutoffs for MTRasym. RESULTS The mean MTRasym values were 0.005 ± 0.005 in the PML group, 0.025 ± 0.005 in the PCNSL group, and 0.025 ± 0.009 in the GBM group. There were significant differences in MTRasym between PML and PCNSL (P = 0.023), and between PML and GBM (P = 0.015). For differentiating PML from PCNSL, an MTRasym threshold of 0.0165 gave diagnostic sensitivity, specificity, positive predictive value, and negative predictive value of 100% (all). For differentiating PML from GBM, an MTRasym threshold of 0.015 gave diagnostic sensitivity, specificity, positive predictive value, and negative predictive value of 100%, 90.9%, 80.0%, and 100%, respectively. CONCLUSION MTRasym values obtained from APT imaging allowed patients with PML to be clearly discriminated from patients with PCNSL or GBM.
Collapse
Affiliation(s)
- Hirofumi Koike
- grid.174567.60000 0000 8902 2273Department of Radiology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Minoru Morikawa
- grid.411873.80000 0004 0616 1585Department of Radiology, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Hideki Ishimaru
- grid.411873.80000 0004 0616 1585Department of Radiology, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Reiko Ideguchi
- grid.174567.60000 0000 8902 2273Department of Radioisotope Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8588 Japan
| | - Masataka Uetani
- grid.174567.60000 0000 8902 2273Department of Radiology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Takeshi Hiu
- grid.174567.60000 0000 8902 2273Department of Neurosurgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Takayuki Matsuo
- grid.174567.60000 0000 8902 2273Department of Neurosurgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Mitsuharu Miyoshi
- grid.481637.f0000 0004 0377 9208MR Application and Workflow, GE Healthcare Japan, 4-7-127 Asahigaoka, Hino, Tokyo 191-8503 Japan
| |
Collapse
|
5
|
Zhou J, Zaiss M, Knutsson L, Sun PZ, Ahn SS, Aime S, Bachert P, Blakeley JO, Cai K, Chappell MA, Chen M, Gochberg DF, Goerke S, Heo HY, Jiang S, Jin T, Kim SG, Laterra J, Paech D, Pagel MD, Park JE, Reddy R, Sakata A, Sartoretti-Schefer S, Sherry AD, Smith SA, Stanisz GJ, Sundgren PC, Togao O, Vandsburger M, Wen Z, Wu Y, Zhang Y, Zhu W, Zu Z, van Zijl PCM. Review and consensus recommendations on clinical APT-weighted imaging approaches at 3T: Application to brain tumors. Magn Reson Med 2022; 88:546-574. [PMID: 35452155 PMCID: PMC9321891 DOI: 10.1002/mrm.29241] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/26/2022] [Accepted: 03/02/2022] [Indexed: 12/16/2022]
Abstract
Amide proton transfer-weighted (APTw) MR imaging shows promise as a biomarker of brain tumor status. Currently used APTw MRI pulse sequences and protocols vary substantially among different institutes, and there are no agreed-on standards in the imaging community. Therefore, the results acquired from different research centers are difficult to compare, which hampers uniform clinical application and interpretation. This paper reviews current clinical APTw imaging approaches and provides a rationale for optimized APTw brain tumor imaging at 3 T, including specific recommendations for pulse sequences, acquisition protocols, and data processing methods. We expect that these consensus recommendations will become the first broadly accepted guidelines for APTw imaging of brain tumors on 3 T MRI systems from different vendors. This will allow more medical centers to use the same or comparable APTw MRI techniques for the detection, characterization, and monitoring of brain tumors, enabling multi-center trials in larger patient cohorts and, ultimately, routine clinical use.
Collapse
Affiliation(s)
- Jinyuan Zhou
- Division of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Moritz Zaiss
- Magnetic Resonance Center, Max Planck Institute for Biological Cybernetics, Tübingen, Germany.,Institute of Neuroradiology, University Hospital Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Linda Knutsson
- Division of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Medical Radiation Physics, Lund University, Lund, Sweden.,F.M. Kirby Research Center for Functional Brain Imaging, Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
| | - Phillip Zhe Sun
- Yerkes Imaging Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Sung Soo Ahn
- Department of Radiology and Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Silvio Aime
- Molecular Imaging Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Peter Bachert
- Department of Medical Physics in Radiology, German Cancer Research Center, Heidelberg, Germany.,Faculty of Physics and Astronomy, University of Heidelberg, Heidelberg, Germany
| | - Jaishri O Blakeley
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kejia Cai
- Department of Radiology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Michael A Chappell
- Mental Health and Clinical Neurosciences and Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, UK.,Nottingham Biomedical Research Centre, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Min Chen
- Department of Radiology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Daniel F Gochberg
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Physics, Vanderbilt University, Nashville, Tennessee, USA
| | - Steffen Goerke
- Department of Medical Physics in Radiology, German Cancer Research Center, Heidelberg, Germany
| | - Hye-Young Heo
- Division of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shanshan Jiang
- Division of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tao Jin
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Seong-Gi Kim
- Center for Neuroscience Imaging Research, Institute for Basic Science and Department of Biomedical Engineering, Sungkyunkwan University, Suwon, South Korea
| | - John Laterra
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
| | - Daniel Paech
- Department of Radiology, German Cancer Research Center, Heidelberg, Germany.,Clinic for Neuroradiology, University Hospital Bonn, Bonn, Germany
| | - Mark D Pagel
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Ravinder Reddy
- Center for Advance Metabolic Imaging in Precision Medicine, Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Akihiko Sakata
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - A Dean Sherry
- Advanced Imaging Research Center and Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas, USA
| | - Seth A Smith
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Greg J Stanisz
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Pia C Sundgren
- Department of Diagnostic Radiology/Clinical Sciences Lund, Lund University, Lund, Sweden.,Lund University Bioimaging Center, Lund University, Lund, Sweden.,Department of Medical Imaging and Physiology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Osamu Togao
- Department of Molecular Imaging and Diagnosis, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Zhibo Wen
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yin Wu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 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, Zhejiang, China
| | - Wenzhen Zhu
- Department of Radiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhongliang Zu
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Peter C M van Zijl
- 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, Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
| |
Collapse
|
6
|
Evaluation of Temozolomide Treatment for Glioblastoma Using Amide Proton Transfer Imaging and Diffusion MRI. Cancers (Basel) 2022; 14:cancers14081907. [PMID: 35454814 PMCID: PMC9031574 DOI: 10.3390/cancers14081907] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/06/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Glioblastoma (GBM), the most frequent and malignant histological type of glioma, is associated with a very high mortality rate. MRI is a useful method for the evaluation of tumor growth. However, there are few studies that have quantitatively evaluated the changes in disease state after TMZ treatment against GBM, and it is not fully understood how the effects of treatment are reflected in the quantitative values measured on MRI. We used the C6 glioma rat model to evaluate the tumor changes due to chemotherapy at different doses of TMZ in terms of quantitative values measured by neurite orientation dispersion and density imaging (NODDI) and amide proton transfer (APT) imaging using 7T-MRI. These methods can evaluate the microstructural changes caused by TMZ-induced tumor growth inhibition. Abstract This study aimed to evaluate tumor changes due to chemotherapy with temozolomide (TMZ) in terms of quantitative values measured by APT imaging and NODDI. We performed TMZ treatment (administered orally by gavage to the TMZ-40 mg and TMZ-60 mg groups) on 7-week-old male Wistar rats with rat glioma C6 implanted in the right brain. T2WI, APT imaging, diffusion tensor imaging (DTI), and NODDI were performed on days 7 and 14 after implantation using 7T-MRI, and the calculated quantitative values were statistically compared. Then, HE staining was performed on brain tissue at day 7 and day 14 for each group to compare the results with the MR images. TMZ treatment inhibited tumor growth and necrotic area formation. The necrotic areas observed upon hematoxylin and eosin (HE) staining were consistent with the MTR low-signal areas observed upon APT imaging. The intracellular volume fraction (ICVF) map of the NODDI could best show the microstructure of the tumor, and its value could significantly highlight the difference in treatment effects at different TMZ doses. APT imaging and NODDI can be used to detect the microstructural changes caused by TMZ-induced tumor growth inhibition. The ICVF may be useful as a parameter for determining the effect of TMZ.
Collapse
|
7
|
Huang J, Chen Z, Park SW, Lai JHC, Chan KWY. Molecular Imaging of Brain Tumors and Drug Delivery Using CEST MRI: Promises and Challenges. Pharmaceutics 2022; 14:451. [PMID: 35214183 PMCID: PMC8880023 DOI: 10.3390/pharmaceutics14020451] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/10/2022] Open
Abstract
Chemical exchange saturation transfer (CEST) magnetic resonance imaging (MRI) detects molecules in their natural forms in a sensitive and non-invasive manner. This makes it a robust approach to assess brain tumors and related molecular alterations using endogenous molecules, such as proteins/peptides, and drugs approved for clinical use. In this review, we will discuss the promises of CEST MRI in the identification of tumors, tumor grading, detecting molecular alterations related to isocitrate dehydrogenase (IDH) and O-6-methylguanine-DNA methyltransferase (MGMT), assessment of treatment effects, and using multiple contrasts of CEST to develop theranostic approaches for cancer treatments. Promising applications include (i) using the CEST contrast of amide protons of proteins/peptides to detect brain tumors, such as glioblastoma multiforme (GBM) and low-grade gliomas; (ii) using multiple CEST contrasts for tumor stratification, and (iii) evaluation of the efficacy of drug delivery without the need of metallic or radioactive labels. These promising applications have raised enthusiasm, however, the use of CEST MRI is not trivial. CEST contrast depends on the pulse sequences, saturation parameters, methods used to analyze the CEST spectrum (i.e., Z-spectrum), and, importantly, how to interpret changes in CEST contrast and related molecular alterations in the brain. Emerging pulse sequence designs and data analysis approaches, including those assisted with deep learning, have enhanced the capability of CEST MRI in detecting molecules in brain tumors. CEST has become a specific marker for tumor grading and has the potential for prognosis and theranostics in brain tumors. With increasing understanding of the technical aspects and associated molecular alterations detected by CEST MRI, this young field is expected to have wide clinical applications in the near future.
Collapse
Affiliation(s)
- Jianpan Huang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (J.H.); (Z.C.); (S.-W.P.); (J.H.C.L.)
| | - Zilin Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (J.H.); (Z.C.); (S.-W.P.); (J.H.C.L.)
| | - Se-Weon Park
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (J.H.); (Z.C.); (S.-W.P.); (J.H.C.L.)
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, China
| | - Joseph H. C. Lai
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (J.H.); (Z.C.); (S.-W.P.); (J.H.C.L.)
| | - Kannie W. Y. Chan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (J.H.); (Z.C.); (S.-W.P.); (J.H.C.L.)
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, China
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518057, China
- Tung Biomedical Science Centre, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
8
|
Zhang H, Zhou J, Peng Y. Amide Proton Transfer-Weighted MR Imaging of Pediatric Central Nervous System Diseases. Magn Reson Imaging Clin N Am 2021; 29:631-641. [PMID: 34717850 DOI: 10.1016/j.mric.2021.06.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Amide proton transfer-weighted (APTw) imaging is a molecular MR imaging technique that can detect the concentration of the amide protons in mobile cellular proteins and peptides or a pH change in vivo. Previous studies have indicated that APTw MR imaging can be used to detect malignant brain tumors, stroke, and other neurologic diseases, although the clinical application in pediatric patients remains limited. The authors briefly introduce the basic principles of APTw imaging. Then, they review early clinical applications of this approach to pediatric central nervous system diseases, including pediatric brain development, hypoxic-ischemic encephalopathy, intracranial infection, and brain tumors.
Collapse
Affiliation(s)
- Hong Zhang
- Department of Radiology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 56 Nan Li Shi Road, Xi Cheng District, Beijing, 100045, China
| | - Jinyuan Zhou
- Division of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Park 336, Baltimore, MD 21287, USA
| | - Yun Peng
- Department of Radiology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 56 Nan Li Shi Road, Xi Cheng District, Beijing, 100045, China.
| |
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW This review aims to cover current MRI techniques for assessing treatment response in brain tumors, with a focus on radio-induced lesions. RECENT FINDINGS Pseudoprogression and radionecrosis are common radiological entities after brain tumor irradiation and are difficult to distinguish from real progression, with major consequences on daily patient care. To date, shortcomings of conventional MRI have been largely recognized but morphological sequences are still used in official response assessment criteria. Several complementary advanced techniques have been proposed but none of them have been validated, hampering their clinical use. Among advanced MRI, brain perfusion measures increase diagnostic accuracy, especially when added with spectroscopy and susceptibility-weighted imaging. However, lack of reproducibility, because of several hard-to-control variables, is still a major limitation for their standardization in routine protocols. Amide Proton Transfer is an emerging molecular imaging technique that promises to offer new metrics by indirectly quantifying intracellular mobile proteins and peptide concentration. Preliminary studies suggest that this noncontrast sequence may add key biomarkers in tumor evaluation, especially in posttherapeutic settings. SUMMARY Benefits and pitfalls of conventional and advanced imaging on posttreatment assessment are discussed and the potential added value of APT in this clinicoradiological evolving scenario is introduced.
Collapse
Affiliation(s)
- Lucia Nichelli
- Department of Neuroradiology, Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Pitié-Salpêtrière-Charles Foix
- Sorbonne Université, INSERM, CNRS, Assistance Publique-Hôpitaux de Paris, Institut du Cerveau et de la Moelle épinière, boulevard de l’Hôpital, Paris
| | - Stefano Casagranda
- Department of Research & Innovation, Olea Medical, avenue des Sorbiers, La Ciotat, France
| |
Collapse
|
10
|
Chan RW, Lawrence LSP, Oglesby RT, Chen H, Stewart J, Theriault A, Campbell M, Ruschin M, Myrehaug S, Atenafu EG, Keller B, Chugh B, MacKenzie S, Tseng CL, Detsky J, Maralani PJ, Czarnota GJ, Stanisz GJ, Sahgal A, Lau AZ. Chemical exchange saturation transfer MRI in central nervous system tumours on a 1.5 T MR-Linac. Radiother Oncol 2021; 162:140-149. [PMID: 34280403 DOI: 10.1016/j.radonc.2021.07.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE To describe the implementation and initial results of using Chemical Exchange Saturation Transfer (CEST) for monitoring patients with central nervous system (CNS) tumours treated using a 1.5 tesla MR-guided radiotherapy system. METHODS CNS patients were treated with up to 30 fractions (total dose up to 60 Gy) using a 1.5 T Elekta Unity MR-Linac. CEST scans were obtained in 54 subjects at one or more time points during treatment. CEST metrics, including the amide magnetization transfer ratio (MTRAmide), nuclear Overhauser effect (NOE) MTR (MTRNOE) and asymmetry, were quantified in phantoms and CNS patients. The signal was investigated between tumour and white matter, across time, and across disease categories including high- and low-grade tumours. RESULTS The gross tumour volume (GTV) exhibited lower MTRAmide and MTRNOE and higher asymmetry compared to contralateral normal appearing white matter. Signal changes in the GTV during fractionated radiotherapy were observed. There were differences between high- and low-grade tumours, with higher CEST asymmetry associated with higher grade disease. CONCLUSION CEST MRI using a 1.5 T MR-Linac was demonstrated to be feasible for in vivo imaging of CNS tumours. CEST images showed tumour/white-matter contrast, temporal CEST signal changes, and associations with tumour grade. These results show promise for the eventual goal of using metabolic imaging to inform the design of adaptive radiotherapy protocols.
Collapse
Affiliation(s)
- Rachel W Chan
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada.
| | - Liam S P Lawrence
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Ryan T Oglesby
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Hanbo Chen
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - James Stewart
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Aimee Theriault
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Mikki Campbell
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Mark Ruschin
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Sten Myrehaug
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Eshetu G Atenafu
- Department of Biostatistics, University Health Network, University of Toronto, Toronto, Canada
| | - Brian Keller
- Department of Biostatistics, University Health Network, University of Toronto, Toronto, Canada
| | - Brige Chugh
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada; Department of Physics, Ryerson University, Toronto, Canada
| | - Scott MacKenzie
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Chia-Lin Tseng
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Jay Detsky
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Pejman J Maralani
- Department of Medical Imaging, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Greg J Czarnota
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada; Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Greg J Stanisz
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada; Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, Lublin, Poland
| | - Arjun Sahgal
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Angus Z Lau
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|
11
|
von Knebel Doeberitz N, Maksimovic S, Loi L, Paech D. [Chemical exchange saturation transfer (CEST) : Magnetic resonance imaging in diagnostic oncology]. Radiologe 2021; 61:43-51. [PMID: 33337509 DOI: 10.1007/s00117-020-00786-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Contrast generation by chemical exchange saturation transfer (CEST) is a recently emerging magnetic resonance imaging (MRI) research field with high clinical potential. METHODS This review covers the methodological principles and summarizes the clinical experience of CEST imaging studies in diagnostic oncology performed to date. RESULTS AND CONCLUSION CEST enables the detection of lowly concentrated metabolites, such as peptides and glucose, through selective saturation of metabolite-bound protons and subsequent magnetization transfer to free water. This technology yields additional information about metabolic activity and the tissue microenvironment without the need for conventional contrast agents or radioactive tracers. Various studies, mainly conducted in patients with neuro-oncolgic diseases, suggest that this technology may aid to assess tumor malignancy as well as therapeutic response prior to and in the first follow-up after intervention. KEY POINTS CEST-MRI enables the indirect detection of metabolites without radioactive tracers or contrast agents. Clinical experience exists especially in the setting of neuro-oncologic imaging. In oncologic imaging, CEST-MRI may improve assessment of prognosis and therapy response.
Collapse
Affiliation(s)
- N von Knebel Doeberitz
- Abteilung Radiologie, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Deutschland
| | - S Maksimovic
- Abteilung Radiologie, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Deutschland
| | - L Loi
- Abteilung Radiologie, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Deutschland
| | - D Paech
- Abteilung Radiologie, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Deutschland.
| |
Collapse
|
12
|
Abdalla G, Hammam A, Anjari M, D'Arco DF, Bisdas DS. Glioma surveillance imaging: current strategies, shortcomings, challenges and outlook. BJR Open 2020; 2:20200009. [PMID: 33178973 PMCID: PMC7594888 DOI: 10.1259/bjro.20200009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 01/11/2023] Open
Abstract
Inaccurate assessment of surveillance imaging to assess response to glioma therapy may have life-changing consequences. Varied management plans including chemotherapy, radiotherapy or immunotherapy may all contribute to heterogeneous post-treatment appearances and the overlap between the morphological features of pseudoprogression, pseudoresponse and radiation necrosis can make their discrimination very challenging. Therefore, there has been a drive to develop objective strategies for post-treatment assessment of brain gliomas. This review discusses the most important of these approaches such as the RANO "Response Assessment in Neuro-Oncology", iRANO "Immunotherapy Response Assessment in Neuro-Oncology" and RAPNO "Response Assessment in Paediatric Neuro-Oncology" models. In addition to these systematic approaches for glioma surveillance, the relatively limited information provided by conventional imaging modalities alone has motivated the development of novel advanced magnetic resonance (MR) and metabolic imaging methods for further discrimination between viable tumour and treatment induced changes. Multiple clinical trials and meta-analyses have investigated the diagnostic performance of these novel techniques in the follow up of brain gliomas, including both single modality descriptive studies and comparative imaging assessment. In this manuscript, we review the literature and discuss the promises and pitfalls of frequently studied modalities in glioma surveillance imaging, including MR perfusion, MR diffusion and MR spectroscopy. In addition, we evaluate other promising MR techniques such as chemical exchange saturation transfer as well as fludeoxyglucose and non-FDG positron emission tomography techniques.
Collapse
Affiliation(s)
- Gehad Abdalla
- Department of Neuroradiology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Trust, London, UK
| | - Ahmed Hammam
- Department of Neuroradiology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Trust, London, UK
| | - Mustafa Anjari
- Department of Neuroradiology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Trust, London, UK
| | - Dr. Felice D'Arco
- Department of Neuroradiology, Great Ormond Street Hospital for Children, London, UK
| | | |
Collapse
|
13
|
Liposomal Lapatinib in Combination with Low-Dose Photodynamic Therapy for the Treatment of Glioma. J Clin Med 2019; 8:jcm8122214. [PMID: 31847378 PMCID: PMC6947404 DOI: 10.3390/jcm8122214] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/06/2019] [Accepted: 12/12/2019] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Malignant gliomas are highly invasive and extremely difficult to treat tumours with poor prognosis and outcomes. Photodynamic therapy (PDT), mediated by Gleolan®, has been studied previously with partial success in treating these tumours and extending lifetime. We aim to determine whether combining PDT using ALA-protoporphyrin IX (PpIX) with a liposomal formulation of the clinical epidermal growth factor receptor (EGFR) inhibitor, lapatinib, would increase the anti-tumour PDT efficacy. METHODS Lapatinib was given in vitro and in vivo 24 h prior to PDT and for 3-5 days following PDT to elicit whether the combination provided any benefits to PDT therapy. Live-cell imaging, in vitro PDT, and in vivo studies were performed to elucidate the effect lapatinib had on PDT for a variety of glioma cell lines and as well as GSC-30 neurospheres in vivo. RESULTS PDT combined with lapatinib led to a significant increase in PpIX accumulation, and reductions in the LD50 of PpIX mediated PDT in two EGFR-driven cell lines, U87 and U87vIII, tested (p < 0.05). PDT + lapatinib elicited stronger MRI-quantified glioma responses following PDT for two human glioma-derived tumours (U87 and GSC-30) in vivo (p < 0.05). Furthermore, PDT leads to enhanced survival in rats following treatment with lapatinib compared to lapatinib alone and PDT alone (p < 0.05). CONCLUSIONS As lapatinib is approved for other oncological indications, a realization of its potential combination with PDT and in fluorescence-guided resection could be readily tested clinically. Furthermore, as its use would only be in acute settings, long-term resistance should not pose an issue as compared to its use as monotherapy.
Collapse
|
14
|
Heo HY, Xu X, Jiang S, Zhao Y, Keupp J, Redmond KJ, Laterra J, van Zijl PC, Zhou J. Prospective acceleration of parallel RF transmission-based 3D chemical exchange saturation transfer imaging with compressed sensing. Magn Reson Med 2019; 82:1812-1821. [PMID: 31209938 PMCID: PMC6660350 DOI: 10.1002/mrm.27875] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/07/2019] [Accepted: 05/30/2019] [Indexed: 12/26/2022]
Abstract
PURPOSE To develop prospectively accelerated 3D CEST imaging using compressed sensing (CS), combined with a saturation scheme based on time-interleaved parallel transmission. METHODS A variable density pseudo-random sampling pattern with a centric elliptical k-space ordering was used for CS acceleration in 3D. Retrospective CS studies were performed with CEST phantoms to test the reconstruction scheme. Prospectively CS-accelerated 3D-CEST images were acquired in 10 healthy volunteers and 6 brain tumor patients with an acceleration factor (RCS ) of 4 and compared with conventional SENSE reconstructed images. Amide proton transfer weighted (APTw) signals under varied RF saturation powers were compared with varied acceleration factors. RESULTS The APTw signals obtained from the CS with acceleration factor of 4 were well-preserved as compared with the reference image (SENSE R = 2) both in retrospective phantom and prospective healthy volunteer studies. In the patient study, the APTw signals were significantly higher in the tumor region (gadolinium [Gd]-enhancing tumor core) than in the normal tissue (p < .001). There was no significant APTw difference between the CS-accelerated images and the reference image. The scan time of CS-accelerated 3D APTw imaging was dramatically reduced to 2:10 minutes (in-plane spatial resolution of 1.8 × 1.8 mm2 ; 15 slices with 4-mm slice thickness) as compared with SENSE (4:07 minutes). CONCLUSION Compressed sensing acceleration was successfully extended to 3D-CEST imaging without compromising CEST image quality and quantification. The CS-based CEST imaging can easily be integrated into clinical protocols and would be beneficial for a wide range of applications.
Collapse
Affiliation(s)
- Hye-Young Heo
- Divison 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
| | - Xiang Xu
- Divison 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
| | - Shanshan Jiang
- Divison of MR Research, Department of Radiology, Johns
Hopkins University, Baltimore, Maryland, USA
| | | | | | - Kristin J. Redmond
- Department of Radiation Oncology and Molecular Radiation
Science, Johns Hopkins University, Baltimore, Maryland, USA
| | - John Laterra
- F.M. Kirby Research Center for Functional Brain Imaging,
Kennedy Krieger Institute, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University,
Baltimore, Maryland, USA
| | - Peter C.M. van Zijl
- Divison 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
- Divison 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
|
15
|
Zhou J, Heo HY, Knutsson L, van Zijl PCM, Jiang S. APT-weighted MRI: Techniques, current neuro applications, and challenging issues. J Magn Reson Imaging 2019; 50:347-364. [PMID: 30663162 DOI: 10.1002/jmri.26645] [Citation(s) in RCA: 252] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 12/26/2018] [Accepted: 12/27/2018] [Indexed: 02/06/2023] Open
Abstract
Amide proton transfer-weighted (APTw) imaging is a molecular MRI technique that generates image contrast based predominantly on the amide protons in mobile cellular proteins and peptides that are endogenous in tissue. This technique, the most studied type of chemical exchange saturation transfer imaging, has been used successfully for imaging of protein content and pH, the latter being possible due to the strong dependence of the amide proton exchange rate on pH. In this article we briefly review the basic principles and recent technical advances of APTw imaging, which is showing promise clinically, especially for characterizing brain tumors and distinguishing recurrent tumor from treatment effects. Early applications of this approach to stroke, Alzheimer's disease, Parkinson's disease, multiple sclerosis, and traumatic brain injury are also illustrated. Finally, we outline the technical challenges for clinical APT-based imaging and discuss several controversies regarding the origin of APTw imaging signals in vivo. Level of Evidence: 3 Technical Efficacy Stage: 3 J. Magn. Reson. Imaging 2019;50:347-364.
Collapse
Affiliation(s)
- Jinyuan Zhou
- 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
| | - 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
| | - Linda Knutsson
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Medical Radiation Physics, Lund University, Lund, Sweden
| | - Peter C M 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
| | - Shanshan Jiang
- 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
|
16
|
Vanhove C, Goethals I. Magnetic resonance imaging-guided radiation therapy using animal models of glioblastoma. Br J Radiol 2018; 92:20180713. [PMID: 30563357 DOI: 10.1259/bjr.20180713] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Glioblastoma is the most aggressive and most common malignant primary brain tumour in adults and has a high mortality and morbidity. Because local tumour control in glioblastoma patients is still elusive in the majority of patients, there is an urgent need for alternative treatment strategies. However, to implement changes to the existing clinical standard of care, research must be conducted to develop alternative treatment strategies. A novel approach in radiotherapy is the introduction of pre-clinical precision image-guided radiation research platforms. The aim of this review is to give a brief overview of the efforts that have been made in the field of radiation research using animal models of glioblastoma. Because MRI has become the reference imaging technique for treatment planning and assessment of therapeutic responses in glioblastoma patients, we will focus in this review on small animal radiotherapy combined with MRI.
Collapse
Affiliation(s)
- Christian Vanhove
- 1 Department of Electronics and Information Systems, Institute Biomedical Technology (IBiTech), Ghent University , Ghent , Belgium
| | - Ingeborg Goethals
- 2 Department of Nuclear Medicine, Ghent University Hospital , Ghent , Belgium
| |
Collapse
|
17
|
Qamar S, King AD, Ai QY, Law BKH, Chan JSM, Poon DMC, Tong M, Mo FKF, Chen W, Bhatia KS, Ahuja AT, Ma BBY, Yeung DKW, Wang YX, Yuan J. Amide proton transfer MRI detects early changes in nasopharyngeal carcinoma: providing a potential imaging marker for treatment response. Eur Arch Otorhinolaryngol 2018; 276:505-512. [DOI: 10.1007/s00405-018-5231-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 11/28/2018] [Indexed: 01/05/2023]
|
18
|
Buck J, Larkin JR, Simard MA, Khrapitchev AA, Chappell MA, Sibson NR. Sensitivity of Multiphase Pseudocontinuous Arterial Spin Labelling (MP pCASL) Magnetic Resonance Imaging for Measuring Brain and Tumour Blood Flow in Mice. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:4580919. [PMID: 30532663 PMCID: PMC6247770 DOI: 10.1155/2018/4580919] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/28/2018] [Accepted: 09/26/2018] [Indexed: 11/17/2022]
Abstract
Brain and tumour blood flow can be measured noninvasively using arterial spin labelling (ASL) magnetic resonance imaging (MRI), but reliable quantification in mouse models remains difficult. Pseudocontinuous ASL (pCASL) is recommended as the clinical standard for ASL and can be improved using multiphase labelling (MP pCASL). The aim of this study was to optimise and validate MP pCASL MRI for cerebral blood flow (CBF) measurement in mice and to assess its sensitivity to tumour perfusion. Following optimization of the MP pCASL sequence, CBF data were compared with gold-standard autoradiography, showing close agreement. Subsequently, MP pCASL data were acquired at weekly intervals in models of primary and secondary brain tumours, and tumour microvessel density was determined histologically. MP pCASL measurements in a secondary brain tumour model revealed a significant reduction in blood flow at day 35 after induction, despite a higher density of blood vessels. Tumour core regions also showed reduced blood flow compared with the tumour rim. Similarly, significant reductions in CBF were found in a model of glioma 28 days after tumour induction, together with an increased density of blood vessels. These findings indicate that MP pCASL MRI provides accurate and robust measurements of cerebral blood flow in naïve mice and is sensitive to changes in tumour perfusion.
Collapse
Affiliation(s)
- Jessica Buck
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, OX3 7LE, Oxford, UK
| | - James R. Larkin
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, OX3 7LE, Oxford, UK
| | - Manon A. Simard
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, OX3 7LE, Oxford, UK
| | - Alexandre A. Khrapitchev
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, OX3 7LE, Oxford, UK
| | - Michael A. Chappell
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, Oxford, UK
| | - Nicola R. Sibson
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, OX3 7LE, Oxford, UK
| |
Collapse
|
19
|
Jiang S, Eberhart CG, Lim M, Heo HY, Zhang Y, Blair L, Wen Z, Holdhoff M, Lin D, Huang P, Qin H, Quinones-Hinojosa A, Weingart JD, Barker PB, Pomper MG, Laterra J, van Zijl PCM, Blakeley JO, Zhou J. Identifying Recurrent Malignant Glioma after Treatment Using Amide Proton Transfer-Weighted MR Imaging: A Validation Study with Image-Guided Stereotactic Biopsy. Clin Cancer Res 2018; 25:552-561. [PMID: 30366937 DOI: 10.1158/1078-0432.ccr-18-1233] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 09/06/2018] [Accepted: 10/22/2018] [Indexed: 12/13/2022]
Abstract
PURPOSE To quantify the accuracy of amide proton transfer-weighted (APTw) MRI for identifying active glioma after treatment via radiographically guided stereotactic tissue validation.Experimental Design: Twenty-one patients who were referred for surgery for MRI features concerning for tumor progression versus treatment effect underwent preoperative APTw imaging. Stereotactic biopsy samples were taken from regions of interest with varying APTw signal intensities. The relationship between final clinical pathology and the histopathology of each of the 64 specimens was analyzed relative to APTw results. Analysis of confirmed recurrent tumor or treatment effect tissue was used to perform ROC analysis. RESULTS Eighteen of 21 patients had recurrent tumor, and 3 had treatment effect on clinical pathology. In 12 patients, there were multiple histopathologic assignments confirmed within the same tumor. Of the 64 total specimens, 20 specimens were active glioma, 27 mixed active and quiescent glioma, and 17 quiescent/no identifiable tumor. APTw signal intensity and histopathologic assignment, cellularity, and proliferation index had significant positive correlations (R = 0.651, 0.580, and 0.458, respectively; all P < 0.001). ROC analysis with a 1.79% APTw intensity cutoff differentiated active from nonactive tumor (AUC of 0.881) with 85.1% sensitivity and 94.1% specificity. Analysis of clinical pathology showed the mean APTw intensity for each patient had 94.4% sensitivity and 100% positive predictive value for identifying recurrent glioma at this cutoff. CONCLUSIONS APTw imaging hyperintensity may be a marker of active malignant glioma. It is able to distinguish between regions of heterogeneous abnormality on anatomic brain MRI with high sensitivity and specificity.
Collapse
Affiliation(s)
- Shanshan Jiang
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| | | | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| | - Hye-Young Heo
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| | - Yi Zhang
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| | - Lindsay Blair
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland
| | - Zhibo Wen
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Matthias Holdhoff
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Doris Lin
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| | - Peng Huang
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Huamin Qin
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | | | - Jon D Weingart
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| | - Peter B Barker
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| | - Martin G Pomper
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| | - John Laterra
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland.,Department of Oncology, Johns Hopkins University, Baltimore, Maryland.,Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland.,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland
| | - Peter C M van Zijl
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland.,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland
| | | | - Jinyuan Zhou
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland. .,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland
| |
Collapse
|
20
|
Zhu Y, Ramasawmy R, Johnson SP, Taylor V, Gibb A, Pedley RB, Chattopadhyay N, Lythgoe MF, Golay X, Bradley D, Walker-Samuel S. Non-invasive imaging of disrupted protein homeostasis induced by proteasome inhibitor treatment using chemical exchange saturation transfer MRI. Sci Rep 2018; 8:15068. [PMID: 30305717 PMCID: PMC6180115 DOI: 10.1038/s41598-018-33549-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 10/02/2018] [Indexed: 11/09/2022] Open
Abstract
Proteasome inhibitors (PIs) are now standard of care for several cancers, and noninvasive biomarkers of treatment response are critically required for early patient stratification and treatment personalization. The present study evaluated whether chemical exchange (CEST) magnetic resonance imaging (MRI) can provide measurements that can be used as the noninvasive biomarkers of proteasome inhibition, alongside diffusion MRI and relaxometry. The sensitivity of human colorectal carcinoma cells to the PI Ixazomib was assessed via in vitro and in vivo dose-response experiments. Acute in vivo response to Ixazomib was assessed at three dosing concentrations, using CEST MRI (amide, amine, hydroxyl signals), diffusion MRI (ADC) and relaxometry (T1, T2). These responses were further evaluated with the known histological markers for Ixazomib and Bradford assay ex vivo. The CEST signal from amides and amines increased in proportion to Ixazomib dose in colorectal cancer xenografts. The cell lines differed in their sensitivity to Ixazomib, which was reflected in the MRI measurements. A mild stimulation in tumor growth was observed at low Ixazomib doses. Our results identify CEST MRI as a promising method for safely and noninvasively monitoring disrupted tumor protein homeostasis induced by proteasome inhibitor treatment, and for stratifying sensitivity between tumor types.
Collapse
Affiliation(s)
- Yanan Zhu
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, United Kingdom
| | - Rajiv Ramasawmy
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, United Kingdom
| | - Sean Peter Johnson
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, United Kingdom
| | - Valerie Taylor
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, United Kingdom
| | - Alasdair Gibb
- Neuroscience, Physiology & Pharmacology, University College London, London, WC1E 6BT, United Kingdom
| | - R Barbara Pedley
- Cancer Institute, University College London, London, WC1E 6DD, United Kingdom
| | - Nibedita Chattopadhyay
- Cancer Pharmacology, Takeda Pharmaceutical International Corporation, Cambridge, MA, 02139, United States
| | - Mark F Lythgoe
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, United Kingdom
| | - Xavier Golay
- Institute of Neurology, University College London, London, WC1N 3BG, United Kingdom
| | - Daniel Bradley
- Biomedical Imaging Group, Takeda Pharmaceutical International Corporation, Cambridge, MA, 02139, United States
| | - Simon Walker-Samuel
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, United Kingdom.
| |
Collapse
|
21
|
Sasaki T, Moritani T, Belay A, Capizzano AA, Sato SP, Sato Y, Kirby P, Ishitoya S, Oya A, Toda M, Takahashi K. Role of the Apparent Diffusion Coefficient as a Predictor of Tumor Progression in Patients with Chordoma. AJNR Am J Neuroradiol 2018; 39:1316-1321. [PMID: 29724767 DOI: 10.3174/ajnr.a5664] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 03/10/2018] [Indexed: 01/15/2023]
Abstract
BACKGROUND AND PURPOSE Diffusion-weighted imaging may aid in distinguishing aggressive chordoma from nonaggressive chordoma. This study explores the prognostic role of the apparent diffusion coefficient in chordomas. MATERIALS AND METHODS Sixteen patients with residual or recurrent chordoma were divided postoperatively into those with an aggressive tumor, defined as a growing tumor having a doubling time of <1 year, and those with a nonaggressive tumor on follow-up MR images. The ability of the ADC to predict an aggressive tumor phenotype was investigated by receiver operating characteristic analysis. The prognostic role of ADC was assessed using a Kaplan-Meier curve with a log-rank test. RESULTS Seven patients died during a median follow-up of 48 months (range, 4-126 months). Five of these 7 patients were in the aggressive tumor group, and 2 were in the nonaggressive tumor group. The mean ADC was significantly lower in the aggressive tumor group than in the nonaggressive tumor group (P = .002). Receiver operating characteristic analysis showed that a cutoff ADC value of 1.494 × 10-3 × mm2/s could be used to diagnose aggressive tumors with an area under the curve of 0.983 (95% CI, 0.911-1.000), a sensitivity of 1.000 (95% CI, 0.541-1.000), and a specificity of 0.900 (95% CI, 0.555-0.998). Furthermore, a cutoff ADC of ≤1.494 × 10-3 × mm2/s was associated with a significantly worse prognosis (P = .006). CONCLUSIONS Lower ADC values could predict tumor progression in postoperative chordomas.
Collapse
Affiliation(s)
- T Sasaki
- From the Departments of Radiology (T.S., T.M., A.B., A.A.C., S.P.S., Y.S.)
- Asahikawa Medical University (T.S., S.I., A.O., M.T., K.T.), Asahikawa, Hokkaido, Japan
| | - T Moritani
- From the Departments of Radiology (T.S., T.M., A.B., A.A.C., S.P.S., Y.S.)
- Department of Radiology (T.M.), University of Michigan, Ann Arbor, Michigan
| | - A Belay
- From the Departments of Radiology (T.S., T.M., A.B., A.A.C., S.P.S., Y.S.)
| | - A A Capizzano
- From the Departments of Radiology (T.S., T.M., A.B., A.A.C., S.P.S., Y.S.)
| | - S P Sato
- From the Departments of Radiology (T.S., T.M., A.B., A.A.C., S.P.S., Y.S.)
| | - Y Sato
- From the Departments of Radiology (T.S., T.M., A.B., A.A.C., S.P.S., Y.S.)
| | - P Kirby
- Pathology (P.K.), University of Iowa, Iowa City, Iowa
| | - S Ishitoya
- Asahikawa Medical University (T.S., S.I., A.O., M.T., K.T.), Asahikawa, Hokkaido, Japan
| | - A Oya
- Asahikawa Medical University (T.S., S.I., A.O., M.T., K.T.), Asahikawa, Hokkaido, Japan
| | - M Toda
- Asahikawa Medical University (T.S., S.I., A.O., M.T., K.T.), Asahikawa, Hokkaido, Japan
| | - K Takahashi
- Asahikawa Medical University (T.S., S.I., A.O., M.T., K.T.), Asahikawa, Hokkaido, Japan
| |
Collapse
|
22
|
Regnery S, Adeberg S, Dreher C, Oberhollenzer J, Meissner JE, Goerke S, Windschuh J, Deike-Hofmann K, Bickelhaupt S, Zaiss M, Radbruch A, Bendszus M, Wick W, Unterberg A, Rieken S, Debus J, Bachert P, Ladd M, Schlemmer HP, Paech D. Chemical exchange saturation transfer MRI serves as predictor of early progression in glioblastoma patients. Oncotarget 2018; 9:28772-28783. [PMID: 29983895 PMCID: PMC6033360 DOI: 10.18632/oncotarget.25594] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 05/24/2018] [Indexed: 12/03/2022] Open
Abstract
PURPOSE To prospectively investigate chemical exchange saturation transfer (CEST) MRI in glioblastoma patients as predictor of early tumor progression after first-line treatment. EXPERIMENTAL DESIGN Twenty previously untreated glioblastoma patients underwent CEST MRI employing a 7T whole-body scanner. Nuclear Overhauser effect (NOE) as well as amide proton transfer (APT) CEST signals were isolated using Lorentzian difference (LD) analysis and relaxation compensated by the apparent exchange-dependent relaxation rate (AREX) evaluation. Additionally, NOE-weighted asymmetric magnetic transfer ratio (MTRasym) and downfield-NOE-suppressed APT (dns-APT) were calculated. Patient response to consecutive treatment was determined according to the RANO criteria. Mean signal intensities of each contrast in the whole tumor area were compared between early-progressive and stable disease. RESULTS Pre-treatment tumor signal intensity differed significantly regarding responsiveness to first-line therapy in NOE-LD (p = 0.0001), NOE-weighted MTRasym (p = 0.0186) and dns-APT (p = 0.0328) contrasts. Hence, significant prediction of early progression was possible employing NOE-LD (AUC = 0.98, p = 0.0005), NOE-weighted MTRasym (AUC = 0.83, p = 0.0166) and dns-APT (AUC = 0.80, p = 0.0318). The NOE-LD provided the highest sensitivity (91%) and specificity (100%). CONCLUSIONS CEST derived contrasts, particularly NOE-weighted imaging and dns-APT, yielded significant predictors of early progression after fist-line therapy in glioblastoma. Therefore, CEST MRI might be considered as non-invasive tool for customization of treatment in the future.
Collapse
Affiliation(s)
- Sebastian Regnery
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Division of Radiology, Heidelberg, Germany
| | - Sebastian Adeberg
- German Cancer Research Center (DKFZ), HIRO (Heidelberg Institute for Radiation Oncology), Heidelberg, Germany
| | - Constantin Dreher
- German Cancer Research Center (DKFZ), Division of Radiology, Heidelberg, Germany
| | | | - Jan-Eric Meissner
- German Cancer Research Center (DKFZ), Division of Medical Physics in Radiology, Heidelberg, Germany
| | - Steffen Goerke
- German Cancer Research Center (DKFZ), Division of Medical Physics in Radiology, Heidelberg, Germany
| | - Johannes Windschuh
- German Cancer Research Center (DKFZ), Division of Medical Physics in Radiology, Heidelberg, Germany
| | | | | | | | - Alexander Radbruch
- German Cancer Research Center (DKFZ), Division of Radiology, Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Wolfgang Wick
- Department of Neurology, University Hospital Heidelberg, Heidelberg, Germany
| | - Andreas Unterberg
- Department of Neurosurgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Rieken
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter Bachert
- German Cancer Research Center (DKFZ), Division of Medical Physics in Radiology, Heidelberg, Germany
| | - Mark Ladd
- German Cancer Research Center (DKFZ), Division of Medical Physics in Radiology, Heidelberg, Germany
- Faculty of Physics and Astronomy, University of Heidelberg, Heidelberg, Germany
- Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
| | | | - Daniel Paech
- German Cancer Research Center (DKFZ), Division of Radiology, Heidelberg, Germany
| |
Collapse
|
23
|
Zhao J, Chen Y, Zhao Y, Yang S, Chen Z, Wu Y. Assessment of chemoradiotherapy response in glioma with magnetic resonance amide proton transfer imaging in a rodent model. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2017; 2017:541-543. [PMID: 29059929 DOI: 10.1109/embc.2017.8036881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Magnetic resonance amide proton transfer (APT) imaging has proved its potential for detecting tumors and evaluating treatment response by depicting chemical exchange saturation transfer effect between the endogenous protein/peptide amide proton and bulk water. However, conventional asymmetry analysis for APT effect measurement is susceptible to concomitant contributions, particular semisolid magnetic transfer (MT) and nuclear overhauser effect. In this study, dominant saturation transfer (ST) effects, including direct water saturation and MT, were estimated from a sum of two Lorentzian functions. APT effect was then quantified by subtracting the Z-spectrum with the two ST effects. Feasibility of the method in the assessment of glioma therapeutic response was investigated in a rodent model at 3 Tesla.
Collapse
|
24
|
Yoon RG, Kim HS, Koh MJ, Shim WH, Jung SC, Kim SJ, Kim JH. Differentiation of Recurrent Glioblastoma from Delayed Radiation Necrosis by Using Voxel-based Multiparametric Analysis of MR Imaging Data. Radiology 2017; 285:206-213. [PMID: 28535120 DOI: 10.1148/radiol.2017161588] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Purpose To assess a volume-weighted voxel-based multiparametric (MP) clustering method as an imaging biomarker to differentiate recurrent glioblastoma from delayed radiation necrosis. Materials and Methods The institutional review board approved this retrospective study and waived the informed consent requirement. Seventy-five patients with pathologic analysis-confirmed recurrent glioblastoma (n = 42) or radiation necrosis (n = 33) who presented with enlarged contrast material-enhanced lesions at magnetic resonance (MR) imaging after they completed concurrent chemotherapy and radiation therapy were enrolled. The diagnostic performance of the total MP cluster score was determined by using the area under the receiver operating characteristic curve (AUC) with cross-validation and compared with those of single parameter measurements (10% histogram cutoffs of apparent diffusion coefficient [ADC10] or 90% histogram cutoffs of normalized cerebral blood volume and initial time-signal intensity AUC). Results Receiver operating characteristic curve analysis showed that an AUC for differentiating recurrent glioblastoma from delayed radiation necrosis was highest in the total MP cluster score and lowest for ADC10 for both readers. The total MP cluster score had significantly better diagnostic accuracy than any single parameter (corrected P = .001-.039 for reader 1; corrected P = .005-.041 for reader 2). The total MP cluster score was the best predictor of recurrent glioblastoma (cross-validated AUCs, 0.942-0.946 for both readers), with a sensitivity of 95.2% for reader 1 and 97.6% for reader 2. Conclusion Quantitative analysis with volume-weighted voxel-based MP clustering appears to be superior to the use of single imaging parameters to differentiate recurrent glioblastoma from delayed radiation necrosis. © RSNA, 2017 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Ra Gyoung Yoon
- From the Department of Radiology, Catholic Kwandong University College of Medicine, Catholic Kwandong University International St. Mary's Hospital, Incheon, Korea (R.G.Y.); Department of Radiology, Jeju National University Hospital, Jeju, Korea (M.J.G.); Department of Radiology and Research Institute of Radiology (H.S.K., W.H.S., S.C.J., S.J.K.) and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul 138-736, Korea
| | - Ho Sung Kim
- From the Department of Radiology, Catholic Kwandong University College of Medicine, Catholic Kwandong University International St. Mary's Hospital, Incheon, Korea (R.G.Y.); Department of Radiology, Jeju National University Hospital, Jeju, Korea (M.J.G.); Department of Radiology and Research Institute of Radiology (H.S.K., W.H.S., S.C.J., S.J.K.) and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul 138-736, Korea
| | - Myeong Ju Koh
- From the Department of Radiology, Catholic Kwandong University College of Medicine, Catholic Kwandong University International St. Mary's Hospital, Incheon, Korea (R.G.Y.); Department of Radiology, Jeju National University Hospital, Jeju, Korea (M.J.G.); Department of Radiology and Research Institute of Radiology (H.S.K., W.H.S., S.C.J., S.J.K.) and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul 138-736, Korea
| | - Woo Hyun Shim
- From the Department of Radiology, Catholic Kwandong University College of Medicine, Catholic Kwandong University International St. Mary's Hospital, Incheon, Korea (R.G.Y.); Department of Radiology, Jeju National University Hospital, Jeju, Korea (M.J.G.); Department of Radiology and Research Institute of Radiology (H.S.K., W.H.S., S.C.J., S.J.K.) and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul 138-736, Korea
| | - Seung Chai Jung
- From the Department of Radiology, Catholic Kwandong University College of Medicine, Catholic Kwandong University International St. Mary's Hospital, Incheon, Korea (R.G.Y.); Department of Radiology, Jeju National University Hospital, Jeju, Korea (M.J.G.); Department of Radiology and Research Institute of Radiology (H.S.K., W.H.S., S.C.J., S.J.K.) and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul 138-736, Korea
| | - Sang Joon Kim
- From the Department of Radiology, Catholic Kwandong University College of Medicine, Catholic Kwandong University International St. Mary's Hospital, Incheon, Korea (R.G.Y.); Department of Radiology, Jeju National University Hospital, Jeju, Korea (M.J.G.); Department of Radiology and Research Institute of Radiology (H.S.K., W.H.S., S.C.J., S.J.K.) and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul 138-736, Korea
| | - Jeong Hoon Kim
- From the Department of Radiology, Catholic Kwandong University College of Medicine, Catholic Kwandong University International St. Mary's Hospital, Incheon, Korea (R.G.Y.); Department of Radiology, Jeju National University Hospital, Jeju, Korea (M.J.G.); Department of Radiology and Research Institute of Radiology (H.S.K., W.H.S., S.C.J., S.J.K.) and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul 138-736, Korea
| |
Collapse
|
25
|
Wang P, Li J, Diao Q, Lin Y, Zhang J, Li L, Yang G, Fang X, Li X, Chen Y, Zheng L, Lu G. Assessment of glioma response to radiotherapy using 3D pulsed-continuous arterial spin labeling and 3D segmented volume. Eur J Radiol 2016; 85:1987-1992. [PMID: 27776650 DOI: 10.1016/j.ejrad.2016.08.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 08/09/2016] [Accepted: 08/13/2016] [Indexed: 12/01/2022]
Abstract
BACKGROUND Gliomas are the most common primary brain tumors in adults, in some cases, radiotherapy may be the preferred treatment option especially for elderly people who cannot endure surgery. Therefore, it is necessary to evaluate the effects of radiotherapy on glioma. Arterial spin labeling (ASL) is an MR imaging technique that allows for a quantitative determination of cerebral blood flow (CBF) noninvasively. Tumor volume is still an important determinant for evaluating treatment response. The purpose of this study was to investigate the relationship between the tumor perfusion parameters and tumor volume and assess the effects of radiotherapy on glioma using pulsed-continuous arterial spin labeling (pcASL) technique. METHODS 35 patients with gliomas, histologically classified as low-grade group (n=16) and high-grade group (n=19), treated with radiotherapy only or before using other therapies were included in this study. MR examinations, including T1 weighted image and pcASL, were performed before and 4, 8, 12, 16 weeks after radiotherapy. Regional CBF of normal tissue, mean tumor blood flow (TBFmean), maximum tumor blood flow (TBFmax), and tumor volume were evaluated at each time point. Both the percentage change in CBF (CBF ratio), TBFmean (TBFmean ratio), TBFmax (TBFmax ratio) and the percentage change in tumor volume (volume ratio) were calculated using values obtained before and after radiotherapy. The correlation between the volume ratio and CBF ratio, TBFmean ratio, TBFmax ratio was assessed using linear regression analysis and Pearson's correlation. RESULTS The TBFmean and TBFmax of high-grade gliomas were significantly higher than that of low-grade group. In high-grade group, a strong correlation was demonstrated between the tumor volume and the TBFmax before radiotherapy (R2=0.35, rs=0.59, p<0.05). There was also a significant correlation between the TBFmax before radiotherapy and the tumor volume ratio before and 8 weeks after radiotherapy (R2=0.56, rs=-0.74, p<0.05). CONCLUSION The TBFmax measured using pcASL could assess tumoral grade and also could become a potential tool for evaluating the therapeutic effects of radiation.
Collapse
Affiliation(s)
- Peng Wang
- Department of Radiology, Nanjing General Hospital of Nanjing Military Command, PLA, No.305, East Zhongshan Road, Nanjing 210002, Jiangsu, China
| | - Jianrui Li
- Department of Radiology, Nanjing General Hospital of Nanjing Military Command, PLA, No.305, East Zhongshan Road, Nanjing 210002, Jiangsu, China
| | - Qiang Diao
- Department of Radiology, Nanjing General Hospital of Nanjing Military Command, PLA, No.305, East Zhongshan Road, Nanjing 210002, Jiangsu, China
| | - YuanKai Lin
- Department of Radiology, Nanjing General Hospital of Nanjing Military Command, PLA, No.305, East Zhongshan Road, Nanjing 210002, Jiangsu, China
| | - Jun Zhang
- Department of Radiology, Nanjing General Hospital of Nanjing Military Command, PLA, No.305, East Zhongshan Road, Nanjing 210002, Jiangsu, China
| | - Lin Li
- Department of Radiology, Nanjing General Hospital of Nanjing Military Command, PLA, No.305, East Zhongshan Road, Nanjing 210002, Jiangsu, China
| | - Gang Yang
- Department of Radiology, Nanjing General Hospital of Nanjing Military Command, PLA, No.305, East Zhongshan Road, Nanjing 210002, Jiangsu, China
| | - Xiaokun Fang
- Department of Radiology, Nanjing General Hospital of Nanjing Military Command, PLA, No.305, East Zhongshan Road, Nanjing 210002, Jiangsu, China
| | - Xie Li
- Department of Radiology, Nanjing General Hospital of Nanjing Military Command, PLA, No.305, East Zhongshan Road, Nanjing 210002, Jiangsu, China
| | - YingQi Chen
- Department of Radiology, Nanjing General Hospital of Nanjing Military Command, PLA, No.305, East Zhongshan Road, Nanjing 210002, Jiangsu, China
| | - Ling Zheng
- Department of Radiology, Nanjing General Hospital of Nanjing Military Command, PLA, No.305, East Zhongshan Road, Nanjing 210002, Jiangsu, China.
| | - Guangming Lu
- Department of Radiology, Nanjing General Hospital of Nanjing Military Command, PLA, No.305, East Zhongshan Road, Nanjing 210002, Jiangsu, China.
| |
Collapse
|
26
|
Xu YJ, Cui Y, Li HX, Shi WQ, Li FY, Wang JZ, Zeng QS. Noninvasive evaluation of radiation-enhanced glioma cells invasiveness by ultra-high-field (1)H-MRS in vitro. Magn Reson Imaging 2016; 34:1121-7. [PMID: 27215950 DOI: 10.1016/j.mri.2016.05.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/16/2016] [Indexed: 01/30/2023]
Abstract
INTRODUCTION Glioma is the most common type of the primary CNS tumor. Radiotherapy is an important treatment measure after surgery. However, its highly invasive character is the main reason of postoperative recurrence. The aim of the study was to probe the correlation between the invasion ability and the metabolite characteristics of glioma cells at the cellular level after irradiation by using 14.7T high-resolution nuclear proton magnetic resonance spectroscopy ((1)H-MRS). METHODS To determine the matrix metalloproteinase-2 (MMP-2) activity and metabolite ratios of glioma cells after irradiation with different doses of X-rays, U87 and C6 glioma cells were exposed to X-ray irradiation of 0, 1, 5, 10, and 15Gy. After 20h, the perchloric acid (PCA) extraction method was used to evaluate water-soluble metabolites [choline (Cho), creatine (Cr), and N-acetylaspartate (NAA)], and (1)H-MRS patterns and changes in metabolite ratios were observed in vitro by 14.7T high resolution (1)H-MRS. Matrigel invasion assays and gelatin zymography were performed to test the invasion ability of U87 and C6 glioma cells. RESULTS Good MR spectra were obtained from PCA method extracts of U87 and C6 glioma cells. Both radiation-induced MMP-2 activity and the Cho/Cr and Cho/NAA ratios increased after irradiation, and their increase occurred in a dose-dependent manner. The MMP-2 activity and the Cho/Cr and Cho/NAA ratios of glioma cells increased after irradiation up to 10Gy and decreased thereafter. In particular, the Cho/NAA ratio of U87 cells increased from 3.55±0.06 (0Gy) to 9.13±0.30 (10Gy) and then declined to 5.94±0.15 (15Gy). Furthermore, the invasion ability of glioma cells had a strong positive correlation with the Cho/Cr and Cho/NAA ratios. Both the Cho/Cr ratio and the Cho/NAA ratio of U87 glioma cells were highly positively correlated with the number of invading cells in the Matrigel invasion assay. The Pearson's correlation coefficient (r) values of U87 cells were 0.89 (Cho/Cr ratio versus invasion ability) and 0.91 (Cho/NAA ratio versus invasion ability) (P<0.01). C6 cells exhibited similar changes to those of U87 cells. CONCLUSIONS In vitro high-resolution (1)H-MRS is useful for detecting glioma invasiveness at the cellular level.
Collapse
Affiliation(s)
- Yan-Jie Xu
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yi Cui
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Hong-Xia Li
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wen-Qi Shi
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Fu-Yan Li
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jian-Zheng Wang
- Department of Radiotherapy, Qilu Hospital of Shandong University, Jinan, China
| | - Qing-Shi Zeng
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
27
|
Heo HY, Lee DH, Zhang Y, Zhao X, Jiang S, Chen M, Zhou J. Insight into the quantitative metrics of chemical exchange saturation transfer (CEST) imaging. Magn Reson Med 2016; 77:1853-1865. [PMID: 27170222 DOI: 10.1002/mrm.26264] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 04/01/2016] [Accepted: 04/14/2016] [Indexed: 12/25/2022]
Abstract
PURPOSE To evaluate the reliability of four CEST imaging metrics for brain tumors, at varied saturation power levels and magnetic field strengths (3-9.4 Tesla (T)). METHODS A five-pool proton exchange model (free water, semisolid, amide, amine, and NOE-related protons) was used for the simulations. For the in vivo study, eight glioma-bearing rats were scanned at 4.7 T. The CEST ratio (CESTR), CESTR normalized with the reference value (CESTRnr ), inverse Z-spectrum-based (MTRRex ), and apparent exchange-related relaxation (AREX) were compared. RESULTS The simulated CEST signal intensities using MTRRex and AREX were substantially increased at relatively high radiofrequency (RF) saturation powers at 3 T and 4.7 T, whereas CESTR and CESTRnr metrics remained relatively stable. There were tremendously high MTRRex and AREX signals around the water frequency at all field strengths because of the small denominators. In the rat tumor study at 4.7 T, both CESTR and CESTRnr showed clear contrasts in the tumor with respect to the normal tissue across all saturation power levels (0.5-3 μT), whereas the AREX showed negligible to negative insignificant contrasts. CONCLUSIONS CEST metrics must be carefully selected based on the different experimental settings. CESTR and CESTRnr are more reliable at 3 T (a clinical field strength) and 4.7 T. Magn Reson Med 77:1853-1865, 2017. © 2016 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Hye-Young Heo
- Divison 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
| | - Dong-Hoon Lee
- Divison of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yi Zhang
- Divison of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Xuna Zhao
- Divison of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Shanshan Jiang
- Divison of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Min Chen
- Department of Radiology, Beijing Hospital, Beijing, China
| | - Jinyuan Zhou
- Divison 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
|
28
|
Yang C, Lee DH, Mangraviti A, Su L, Zhang K, Zhang Y, Zhang B, Li W, Tyler B, Wong J, Wang KKH, Velarde E, Zhou J, Ding K. Quantitative correlational study of microbubble-enhanced ultrasound imaging and magnetic resonance imaging of glioma and early response to radiotherapy in a rat model. Med Phys 2016; 42:4762-72. [PMID: 26233204 DOI: 10.1118/1.4926550] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Radiotherapy remains a major treatment method for malignant tumors. Magnetic resonance imaging (MRI) is the standard modality for assessing glioma treatment response in the clinic. Compared to MRI, ultrasound imaging is low-cost and portable and can be used during intraoperative procedures. The purpose of this study was to quantitatively compare contrast-enhanced ultrasound (CEUS) imaging and MRI of irradiated gliomas in rats and to determine which quantitative ultrasound imaging parameters can be used for the assessment of early response to radiation in glioma. METHODS Thirteen nude rats with U87 glioma were used. A small thinned skull window preparation was performed to facilitate ultrasound imaging and mimic intraoperative procedures. Both CEUS and MRI with structural, functional, and molecular imaging parameters were performed at preradiation and at 1 day and 4 days postradiation. Statistical analysis was performed to determine the correlations between MRI and CEUS parameters and the changes between pre- and postradiation imaging. RESULTS Area under the curve (AUC) in CEUS showed significant difference between preradiation and 4 days postradiation, along with four MRI parameters, T2, apparent diffusion coefficient, cerebral blood flow, and amide proton transfer-weighted (APTw) (all p < 0.05). The APTw signal was correlated with three CEUS parameters, rise time (r = - 0.527, p < 0.05), time to peak (r = - 0.501, p < 0.05), and perfusion index (r = 458, p < 0.05). Cerebral blood flow was correlated with rise time (r = - 0.589, p < 0.01) and time to peak (r = - 0.543, p < 0.05). CONCLUSIONS MRI can be used for the assessment of radiotherapy treatment response and CEUS with AUC as a new technique and can also be one of the assessment methods for early response to radiation in glioma.
Collapse
Affiliation(s)
- Chen Yang
- Department of Ultrasound, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Dong-Hoon Lee
- Division of MR Research, Department of Radiology, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21287
| | - Antonella Mangraviti
- Department of Neurosurgery, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21287
| | - Lin Su
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21231
| | - Kai Zhang
- Division of MR Research, Department of Radiology, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21287
| | - Yin Zhang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21231
| | - Bin Zhang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21231
| | - Wenxiao Li
- Division of MR Research, Department of Radiology, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21287
| | - Betty Tyler
- Department of Neurosurgery, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21287
| | - John Wong
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21231
| | - Ken Kang-Hsin Wang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21231
| | - Esteban Velarde
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21231
| | - Jinyuan Zhou
- Division of MR Research, Department of Radiology, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21287
| | - Kai Ding
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21231
| |
Collapse
|
29
|
Yu Y, Lee DH, Peng SL, Zhang K, Zhang Y, Jiang S, Zhao X, Heo HY, Wang X, Chen M, Lu H, Li H, Zhou J. Assessment of Glioma Response to Radiotherapy Using Multiple MRI Biomarkers with Manual and Semiautomated Segmentation Algorithms. J Neuroimaging 2016; 26:626-634. [PMID: 27128445 DOI: 10.1111/jon.12354] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/23/2016] [Accepted: 03/28/2016] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Multimodality magnetic resonance imaging (MRI) can provide complementary information in the assessment of brain tumors. We aimed to segment tumor in amide proton transfer-weighted (APTw) images and to investigate multiparametric MRI biomarkers for the assessment of glioma response to radiotherapy. For tumor extraction, we evaluated a semiautomated segmentation method based on region of interest (ROI) results by comparing it with the manual segmentation method. METHODS Thirteen nude rats injected with U87 tumor cells were irradiated by an 8-Gy radiation dose. All MRI scans were performed on a 4.7-T animal scanner preradiation, and at day 1, day 4, and day 8 postradiation. Two experts performed manual and semiautomated methods to extract tumor ROIs on APTw images. Multimodality MRI signals of the tumors, including structural (T2 and T1 ), functional (apparent diffusion coefficient and blood flow), and molecular (APTw and magnetization transfer ratio or MTR), were calculated and compared quantitatively. RESULTS The semiautomated method provided more reliable tumor extraction results on APTw images than the manual segmentation, in less time. A considerable increase in the ADC intensities of the tumor was observed during the postradiation. A steady decrease in the blood flow values and in the APTw signal intensities were found after radiotherapy. CONCLUSIONS The semiautomated method of tumor extraction showed greater efficiency and stability than the manual method. Apparent diffusion coefficient, blood flow, and APTw are all useful biomarkers in assessing glioma response to radiotherapy.
Collapse
Affiliation(s)
- Yang Yu
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, China.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, China
| | - Dong-Hoon Lee
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, China
| | - Shin-Lei Peng
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, China
| | - Kai Zhang
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, China
| | - Yi Zhang
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, China
| | - Shanshan Jiang
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, China
| | - Xuna Zhao
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, China
| | - Hye-Young Heo
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, China
| | - Xiangyang Wang
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, China.,Department of Radiology, Beijing Hospital, Beijing, China
| | - Min Chen
- Department of Radiology, Beijing Hospital, Beijing, China
| | - Hanzhang Lu
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, China
| | - Haiyun Li
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, China
| | - Jinyuan Zhou
- Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, China.
| |
Collapse
|
30
|
Huang J, Zhang M, Lu J, Cai C, Chen L, Cai S. A fast chemical exchange saturation transfer imaging scheme based on single-shot spatiotemporal encoding. Magn Reson Med 2016; 77:1786-1796. [PMID: 27120691 DOI: 10.1002/mrm.26258] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 03/30/2016] [Accepted: 04/01/2016] [Indexed: 01/31/2023]
Abstract
PURPOSE To design a new approach that can not only keep the spatial and temporal resolution but also have better built-in immunity to magnetic field inhomogeneity and chemical shift effects than the single-shot echo planar imaging (EPI) for chemical exchange saturation transfer (CEST) MRI. METHOD The single-shot spatiotemporally encoded (SPEN) MRI sequence was combined with a continuous wave saturation pulse for fast CEST MRI (CEST-SPEN MRI). The resulting images were super-resolved reconstructed by a hybrid method that solves the l1 norm minimization together with total variation (TV) regularization. Partial Lorentzian fitting was used to analyze the subsequent Z-spectra. RESULTS Experimental results of a creatine phantom and in vivo tumor rat brains show that CEST-SPEN MRI has good capability in providing CEST-based and NOE-based contrast images. CONCLUSIONS Compared with CEST-EPI, CEST-SPEN MRI has better immunity to magnetic field inhomogeneity and provides better contrast images within identical acquisition time, especially under an identical inhomogeneous field. Magn Reson Med 77:1786-1796, 2017. © 2016 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Jianpan Huang
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China
| | - Miao Zhang
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China
| | - Jianhua Lu
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China.,Meizhouwan Vocational Technology College, Putian, China
| | - Congbo Cai
- Department of Communication Engineering, Xiamen University, Xiamen, China
| | - Lin Chen
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China
| | - Shuhui Cai
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China
| |
Collapse
|
31
|
Yan K, Fu Z, Yang C, Zhang K, Jiang S, Lee DH, Heo HY, Zhang Y, Cole RN, Van Eyk JE, Zhou J. Assessing Amide Proton Transfer (APT) MRI Contrast Origins in 9 L Gliosarcoma in the Rat Brain Using Proteomic Analysis. Mol Imaging Biol 2016; 17:479-87. [PMID: 25622812 DOI: 10.1007/s11307-015-0828-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE To investigate the biochemical origin of the amide photon transfer (APT)-weighted hyperintensity in brain tumors. PROCEDURES Seven 9 L gliosarcoma-bearing rats were imaged at 4.7 T. Tumor and normal brain tissue samples of equal volumes were prepared with a coronal rat brain matrix and a tissue biopsy punch. The total tissue protein and the cytosolic subproteome were extracted from both samples. Protein samples were analyzed using two-dimensional gel electrophoresis, and the proteins with significant abundance changes were identified by mass spectrometry. RESULTS There was a significant increase in the cytosolic protein concentration in the tumor, compared to normal brain regions, but the total protein concentrations were comparable. The protein profiles of the tumor and normal brain tissue differed significantly. Six cytosolic proteins, four endoplasmic reticulum proteins, and five secreted proteins were considerably upregulated in the tumor. CONCLUSIONS Our experiments confirmed an increase in the cytosolic protein concentration in tumors and identified several key proteins that may cause APT-weighted hyperintensity.
Collapse
Affiliation(s)
- Kun Yan
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Heo HY, Zhang Y, Lee DH, Jiang S, Zhao X, Zhou J. Accelerating chemical exchange saturation transfer (CEST) MRI by combining compressed sensing and sensitivity encoding techniques. Magn Reson Med 2016; 77:779-786. [PMID: 26888295 DOI: 10.1002/mrm.26141] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/02/2016] [Accepted: 01/04/2016] [Indexed: 12/16/2022]
Abstract
PURPOSE To evaluate the feasibility of accelerated chemical-exchange-saturation-transfer (CEST) imaging using a combination of compressed sensing (CS) and sensitivity encoding (SENSE) at 3 Tesla. THEORY AND METHODS Two healthy volunteers and six high-grade glioma patients were recruited. Raw CEST image k-space data were acquired (with varied radiofrequency saturation power levels for the healthy volunteer study), and a sequential CS and SENSE reconstruction (CS-SENSE) was assessed. The MTRasym (3.5 ppm) signals were compared with varied CS-SENSE acceleration factors. RESULTS In the healthy volunteer study, a CS-SENSE acceleration factor of R = 2 × 2 (CS × SENSE) was achieved without compromising the reconstructed MTRasym (3.5 ppm) image quality. The MTRasym (3.5 ppm) signals obtained from the CS-SENSE reconstruction with R = 2 × 2 were well preserved compared with the reference image (R = 2 for only SENSE). In the glioma patient study, the MTRasym (3.5 ppm) signals were significantly higher in the tumor region (Gd-enhancing tumor core) than in the normal-appearing white matter (P < 0.001). There was no significant MTRasym (3.5 ppm) difference between the reference image and CS-SENSE-reconstructed image in the acceleration factor of R = 2 × 2. CONCLUSION Combining the SENSE technique with CS (R = 2 × 2) enables considerable acceleration of CEST image acquisition and potentially has a wide range of clinical applications. Magn Reson Med 77:779-786, 2017. © 2016 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Hye-Young Heo
- Divison 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
| | - Yi Zhang
- Divison of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dong-Hoon Lee
- Divison of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shanshan Jiang
- Divison of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xuna Zhao
- Divison of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jinyuan Zhou
- Divison 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
|
33
|
Added value of amide proton transfer imaging to conventional and perfusion MR imaging for evaluating the treatment response of newly diagnosed glioblastoma. Eur Radiol 2016; 26:4390-4403. [PMID: 26883333 DOI: 10.1007/s00330-016-4261-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 01/20/2016] [Accepted: 02/01/2016] [Indexed: 01/17/2023]
Abstract
OBJECTIVES To determine the added value of amide proton transfer (APT) imaging to conventional and perfusion MRI for differentiating tumour progression (TP) from the treatment-related effect (TE) in patients with post-treatment glioblastomas. METHODS Sixty-five consecutive patients with enlarging contrast-enhancing lesions following concurrent chemoradiotherapy were assessed using contrast-enhanced T1-weighted MRI (CE-T1WI), 90th percentile histogram parameters of normalized cerebral blood volume (nCBV90) and APT asymmetry value (APT90). Diagnostic performance was determined using the area under the receiver operating characteristic curve (AUC) and cross validations. RESULTS There were statistically significant differences in the mean APT90 between the TP and the TE groups (3.87-4.01 % vs. 1.38-1.41 %; P < .001). Compared with CE-T1WI alone, the addition of APT90 to CE-T1WI significantly improved cross-validated AUC from 0.58-0.74 to 0.89-0.91 for differentiating TP from TE. The combination of CE-T1WI, nCBV90 and APT90 resulted in greater diagnostic accuracy for differentiating TP from TE than the combination of CE-T1WI and nCBV90 (cross-validated AUC, 0.95-0.97 vs. 0.84-0.91). The inter-reader agreement between the expert and trainee was excellent for the measurements of APT90 (intraclass correlation coefficient, 0.94). CONCLUSION Adding APT imaging to conventional and perfusion MRI improves the diagnostic performance for differentiating TP from TE. KEY POINTS • APT imaging could provide a reliable distinction between TP and TE • Adding APT imaging to CE-T1WI improved the diagnostic accuracy versus CE-T1WI alone • Multimodal imaging using CE-T1WI, perfusion and APT imaging led to accurate diagnosis • The inter-reader agreement of APT histogram parameters was excellent.
Collapse
|
34
|
Lee DH, Heo HY, Zhang K, Zhang Y, Jiang S, Zhao X, Zhou J. Quantitative assessment of the effects of water proton concentration and water T 1 changes on amide proton transfer (APT) and nuclear overhauser enhancement (NOE) MRI: The origin of the APT imaging signal in brain tumor. Magn Reson Med 2016; 77:855-863. [PMID: 26841096 DOI: 10.1002/mrm.26131] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 12/30/2015] [Accepted: 12/30/2015] [Indexed: 01/02/2023]
Abstract
PURPOSE To quantify pure chemical exchange-dependent saturation transfer (CEST) related amide proton transfer (APT) and nuclear Overhauser enhancement (NOE) signals in a rat glioma model and to investigate the mixed effects of water content and water T1 on APT and NOE imaging signals. METHODS Eleven U87 tumor-bearing rats were scanned at 4.7 T. A relatively accurate mathematical approach, based on extrapolated semisolid magnetization-transfer reference signals, was used to remove the concurrent effects of direct water saturation and semisolid magnetization-transfer. Pure APT and NOE signals, in addition to the commonly used magnetization-transfer-ratio asymmetry at 3.5 ppm, MTRasym (3.5ppm), were assessed. RESULTS The measured APT signal intensity of the tumor (11.06%, much larger than the value reported in the literature) was the major contributor (approximately 80.6%) to the MTRasym (3.5ppm) contrast between the tumor and the contralateral brain region. Both the water content ([water proton]) and water T1 (T1w ) were increased in the tumor, but there were no significant correlations among APT, NOE, or MTRasym (3.5ppm) signals and T1w /[water proton]. CONCLUSION The effect of increasing T1w on the CEST signal in the tumor was mostly eliminated by the effect of increasing water content, and the observed APT-weighted hyperintensity in the tumor should be dominated by the increased amide proton concentration. Magn Reson Med 77:855-863, 2017. © 2016 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Dong-Hoon Lee
- Division of MR Research, Dept. of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hye-Young Heo
- Division of MR Research, Dept. of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kai Zhang
- Division of MR Research, Dept. of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yi Zhang
- Division of MR Research, Dept. of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shanshan Jiang
- Division of MR Research, Dept. of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xuna Zhao
- Division of MR Research, Dept. of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jinyuan Zhou
- Division of MR Research, Dept. 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
|
35
|
Ma B, Blakeley JO, Hong X, Zhang H, Jiang S, Blair L, Zhang Y, Heo HY, Zhang M, van Zijl PCM, Zhou J. Applying amide proton transfer-weighted MRI to distinguish pseudoprogression from true progression in malignant gliomas. J Magn Reson Imaging 2016; 44:456-62. [PMID: 26788865 DOI: 10.1002/jmri.25159] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 01/04/2016] [Indexed: 11/07/2022] Open
Abstract
PURPOSE To assess amide proton transfer-weighted (APTW) imaging features in patients with malignant gliomas after chemoradiation and the diagnostic performance of APT imaging for distinguishing true progression from pseudoprogression. MATERIALS AND METHODS After approval by the Institutional Review Board, 32 patients with clinically suspected tumor progression in the first 3 months after chemoradiation were enrolled and scanned at 3T. Longitudinal routine magnetic resonance imaging (MRI) changes and medical records were assessed to confirm true progression versus pseudoprogression. True progression was defined as lesions progressing on serial imaging over 6 months, and pseudoprogression was defined as lesions stabilizing or regressing without intervention. The APTWmean and APTWmax signals were obtained from three to five regions of interests for each patient and compared between the true progression and pseudoprogression groups. The diagnostic performance was assessed with receiver operating characteristic curve analysis. RESULTS The true progression was associated with APTW hyperintensity (APTWmean = 2.75% ± 0.42%), while pseudoprogression was associated with APTW isointensity to mild hyperintensity (APTWmean = 1.56% ± 0.42%). The APTW signal intensities were significantly higher in the true progression group (n = 20) than in the pseudoprogression group (P < 0.001; n = 12). The cutoff APTWmean and APTWmax intensity values to distinguish between true progression and pseudoprogression were 2.42% (with a sensitivity of 85.0% and a specificity of 100%) and 2.54% (with a sensitivity of 95.0% and a specificity of 91.7%), respectively. CONCLUSION The APTW-MRI signal is a valuable imaging biomarker for distinguishing pseudoprogression from true progression in glioma patients. J. Magn. Reson. Imaging 2016;44:456-462.
Collapse
Affiliation(s)
- Bo Ma
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China.,Department of Radiology, Henan Provincial People's Hospital, Zhengzhou, Henan, PR China
| | - Jaishri O Blakeley
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Xiaohua Hong
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hongyan Zhang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Shanshan Jiang
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Lindsay Blair
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Neurology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yi Zhang
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hye-Young Heo
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Mingzhi Zhang
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Peter C M van Zijl
- 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
- 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
|
36
|
Heo HY, Zhang Y, Jiang S, Lee DH, Zhou J. Quantitative assessment of amide proton transfer (APT) and nuclear overhauser enhancement (NOE) imaging with extrapolated semisolid magnetization transfer reference (EMR) signals: II. Comparison of three EMR models and application to human brain glioma at 3 Tesla. Magn Reson Med 2015; 75:1630-9. [PMID: 26033553 DOI: 10.1002/mrm.25795] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 04/08/2015] [Accepted: 05/01/2015] [Indexed: 12/27/2022]
Abstract
PURPOSE To evaluate the use of three extrapolated semisolid magnetization transfer reference (EMR) methods to quantify amide proton transfer (APT) and nuclear Overhauser enhancement (NOE) signals in human glioma. METHODS Eleven patients with high-grade glioma were scanned at 3 Tesla. aEMR(2) (asymmetric magnetization-transfer or MT model to fit two-sided, wide-offset data), sEMR(2) (symmetric MT model to fit two-sided, wide-offset data), and sEMR(1) (symmetric MT model to fit one-sided, wide-offset data) were assessed. ZEMR and experimental data at 3.5 ppm and -3.5 ppm were subtracted to calculate the APT and NOE signals (APT(#) and NOE(#)), respectively. RESULTS The aEMR(2) and sEMR(1) models provided quite similar APT(#) signals, while the sEMR(2) provided somewhat lower APT(#) signals. The aEMR(2) had an erroneous NOE(#) quantification. Calculated APT(#) signal intensities of glioma (∼4%), much larger than the values reported previously, were significantly higher than those of edema and normal tissue. Compared with normal tissue, gadolinium-enhancing tumor cores were consistently hyperintense on the APT(#) maps and slightly hypointense on the NOE(#) maps. CONCLUSION The sEMR(1) model is the best choice for accurately quantifying APT and NOE signals. The APT-weighted hyperintensity in the tumor was dominated by the APT effect, and the MT asymmetry at 3.5 ppm is a reliable and valid metric for APT imaging of gliomas at 3T.
Collapse
Affiliation(s)
- Hye-Young Heo
- Divison of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yi Zhang
- Divison of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Shanshan Jiang
- Divison of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Dong-Hoon Lee
- Divison of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jinyuan Zhou
- Divison 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
|
37
|
Molecular MRI differentiation between primary central nervous system lymphomas and high-grade gliomas using endogenous protein-based amide proton transfer MR imaging at 3 Tesla. Eur Radiol 2015; 26:64-71. [PMID: 25925361 DOI: 10.1007/s00330-015-3805-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 03/18/2015] [Accepted: 04/14/2015] [Indexed: 01/13/2023]
Abstract
OBJECTIVES To show the ability of using the amide proton transfer-weighted (APTW) MRI signals as imaging biomarkers to differentiate primary central nervous system lymphomas (PCNSLs) from high-grade gliomas (HGGs). METHODS Eleven patients with lymphomas and 21 patients with HGGs were examined. Magnetization-transfer (MT) spectra over an offset range of ± 6 ppm and the conventional MT ratio (MTR) at 15.6 ppm were acquired. The APTW signals, total chemical-exchange-saturation-transfer signal (integral between 0 and 5 ppm, CEST total), and MTR signal were obtained and compared between PCNSLs and HGGs. The diagnostic performance was assessed with the receiver operating characteristic (ROC) curve analysis. RESULTS The PCNSLs usually showed more homogeneous APTW hyperintensity (spatially compared to normal brain tissue) than the HGGs. The APTW max, APTW max-min and CEST total signal intensities were significantly lower (P < 0.05, 0.001 and 0.05, respectively), while the APTW min and MTR were significantly higher (both P < 0.01) in PCNSL lesions than in HGG lesions. The APTW values in peritumoral oedema were significantly lower for PCNSLs than for HGGs (P < 0.01). APTW max-min had the highest area under the ROC curve (0.963) and accuracy (94.1 %) in differentiating PCNSLs from HGGs. CONCLUSIONS The protein-based APTW signal would be a valuable MRI biomarker by which to identify PCNSLs and HGGs presurgically. KEY POINTS PCNSLs overall showed more homogeneous APTW hyperintensity than HGGs. Maximum APTW signals were lower in PCNSL lesions than in HGG lesions. MTR signals were higher in PCNSLs than in HGGs. APTW heterogeneity had the highest accuracy in differentiating PCNSLs from HGGs.
Collapse
|
38
|
Heo HY, Zhang Y, Lee DH, Hong X, Zhou J. Quantitative assessment of amide proton transfer (APT) and nuclear overhauser enhancement (NOE) imaging with extrapolated semi-solid magnetization transfer reference (EMR) signals: Application to a rat glioma model at 4.7 Tesla. Magn Reson Med 2015; 75:137-49. [PMID: 25753614 DOI: 10.1002/mrm.25581] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 11/18/2014] [Accepted: 11/21/2014] [Indexed: 12/15/2022]
Abstract
PURPOSE To quantify amide proton transfer (APT) and nuclear Overhauser enhancement (NOE) contributions to in vivo chemical exchange saturation transfer MRI signals in tumors. THEORY AND METHODS Two-pool (free water and semi-solid protons) and four-pool (free water, semi-solid, amide, and upfield NOE-related protons) tissue models combined with the super-Lorentzian lineshape for semi-solid protons were used to fit wide and narrow frequency-offset magnetization-transfer (MT) data, respectively. Extrapolated semi-solid MT signals at 3.5 and -3.5 ppm from water were used as reference signals to quantify APT and NOE, respectively. Six glioma-bearing rats were scanned at 4.7 Tesla. Quantitative APT and NOE signals were compared at three saturation power levels. RESULTS The observed APT signals were significantly higher in the tumor (center and rim) than in the contralateral normal brain tissue at all saturation powers, and were the major contributor to the APT-weighted image contrast (based on MT asymmetry analysis) between the tumor and the normal brain tissue. The NOE (a positive confounding factor) enhanced this APT-weighted image contrast. The fitted amide pool sizes were significantly larger, while the NOE-related pool sizes were significantly smaller in the tumor than in the normal brain tissue. CONCLUSION The extrapolated semi-solid magnetization transfer reference provides a relatively accurate approach for quantitatively measuring pure APT and NOE signals.
Collapse
Affiliation(s)
- Hye-Young Heo
- Divison of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yi Zhang
- Divison of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dong-Hoon Lee
- Divison of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xiaohua Hong
- Divison of MR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jinyuan Zhou
- Divison 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
|
39
|
Park JE, Kim HS, Goh MJ, Kim SJ, Kim JH. Pseudoprogression in Patients with Glioblastoma: Assessment by Using Volume-weighted Voxel-based Multiparametric Clustering of MR Imaging Data in an Independent Test Set. Radiology 2015; 275:792-802. [PMID: 25611736 DOI: 10.1148/radiol.14141414] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE To validate a volume-weighted voxel-based multiparametric clustering (VVMC) method for magnetic resonance imaging data that is designed to differentiate between pseudoprogression and early tumor progression (ETP) in patients with glioblastoma in an independent test set. MATERIALS AND METHODS This retrospective study was approved by the local institutional review board, with waiver of the need to obtain informed consent. The study patients were grouped chronologically into a training set (108 patients) and a test set (54 patients). The reference standard was pathologic findings or subsequent clinical-radiologic study results. By using the optimal cutoff determined in the training set, the diagnostic performance of VVMC was subsequently tested in the test set and was compared with that of single-parameter measurements (apparent diffusion coefficient [ADC], normalized cerebral blood volume [nCBV], and initial area under the time-signal intensity curve). RESULTS Interreader agreement was highest for VVMC (intraclass correlation coefficient, 0.87-0.89). Receiver operating characteristic curve analysis revealed that VVMC performed the best as a classifier, although statistical significance was not demonstrated with respect to the nCBV in the training set. In the test set, the diagnostic accuracy of VVMC was higher than that of any single-parameter measurements, but this trend reached significance only for the ADC. When the entire population was considered, VVMC had significantly better diagnostic accuracy than did any single parameter (P = .003-.046 for reader 1; P = .002-.016 for reader 2). Results of fivefold cross validation confirmed the trends in both the training set and the test set. CONCLUSION VVMC is a superior and more reproducible imaging biomarker than single-parameter measurements for differentiating between pseudoprogression and ETP in patients with glioblastoma. Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Ji Eun Park
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., M.J.G., S.J.K.) and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul 138-736, Korea
| | | | | | | | | |
Collapse
|
40
|
Sakata A, Okada T, Yamamoto A, Kanagaki M, Fushimi Y, Okada T, Dodo T, Arakawa Y, Schmitt B, Miyamoto S, Togashi K. Grading glial tumors with amide proton transfer MR imaging: different analytical approaches. J Neurooncol 2015; 122:339-48. [PMID: 25559689 DOI: 10.1007/s11060-014-1715-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 12/29/2014] [Indexed: 11/24/2022]
Abstract
Amide proton transfer (APT) magnetic resonance imaging is gaining attention for its capability for grading glial tumors. Usually, a representative slice is analyzed. Different definitions of tumor areas have been employed in previous studies. We hypothesized that the accuracy of APT imaging for brain tumor grading may depend upon the analytical methodology used, such as selection of regions of interest (ROIs), single or multiple tumor slices, and whether or not there is normalization to the contralateral white matter. This study was approved by the institutional review board, and written informed consent was waived. Twenty-six patients with histologically proven glial tumors underwent preoperative APT imaging with a three-dimensional gradient-echo sequence. Two neuroradiologists independently analyzed APT asymmetry (APTasym) images by placing ROIs on both a single representative slice (RS) and all slices including tumor (i.e. whole tumor: WT). ROIs indicating tumor extent were separately defined on both FLAIR and, if applicable, contrast-enhanced T1-weighted images (CE-T1WI), yielding four mean APTasym values (RS-FLAIR, WT-FLAIR, RS-CE-T1WI, and WT-CE-T1WI). The maximum values were also measured using small ROIs, and their differences among grades were evaluated. Receiver operating characteristic (ROC) curve analysis was also conducted on mean and maximum values. Intra-class correlation coefficients for inter-observer agreement were excellent. Significant differences were observed between high- and low-grade gliomas for all five methods (P < 0.01). ROC curve analysis found no statistically significant difference among them. This study clarifies that single-slice APT analysis is robust despite tumor heterogeneity, and can grade glial tumors with or without the use of contrast material.
Collapse
Affiliation(s)
- Akihiko Sakata
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Yeung TPC, Kurdi M, Wang Y, Al-Khazraji B, Morrison L, Hoffman L, Jackson D, Crukley C, Lee TY, Bauman G, Yartsev S. CT perfusion imaging as an early biomarker of differential response to stereotactic radiosurgery in C6 rat gliomas. PLoS One 2014; 9:e109781. [PMID: 25329655 PMCID: PMC4201465 DOI: 10.1371/journal.pone.0109781] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 09/02/2014] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The therapeutic efficacy of stereotactic radiosurgery for glioblastoma is not well understood, and there needs to be an effective biomarker to identify patients who might benefit from this treatment. This study investigated the efficacy of computed tomography (CT) perfusion imaging as an early imaging biomarker of response to stereotactic radiosurgery in a malignant rat glioma model. METHODS Rats with orthotopic C6 glioma tumors received either mock irradiation (controls, N = 8) or stereotactic radiosurgery (N = 25, 12 Gy in one fraction) delivered by Helical Tomotherapy. Twelve irradiated animals were sacrificed four days after stereotactic radiosurgery to assess acute CT perfusion and histological changes, and 13 irradiated animals were used to study survival. Irradiated animals with survival >15 days were designated as responders while those with survival ≤15 days were non-responders. Longitudinal CT perfusion imaging was performed at baseline and regularly for eight weeks post-baseline. RESULTS Early signs of radiation-induced injury were observed on histology. There was an overall survival benefit following stereotactic radiosurgery when compared to the controls (log-rank P<0.04). Responders to stereotactic radiosurgery showed lower relative blood volume (rBV), and permeability-surface area (PS) product on day 7 post-stereotactic radiosurgery when compared to controls and non-responders (P<0.05). rBV and PS on day 7 showed correlations with overall survival (P<0.05), and were predictive of survival with 92% accuracy. CONCLUSIONS Response to stereotactic radiosurgery was heterogeneous, and early selection of responders and non-responders was possible using CT perfusion imaging. Validation of CT perfusion indices for response assessment is necessary before clinical implementation.
Collapse
Affiliation(s)
- Timothy Pok Chi Yeung
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- Robarts Research Institute, Western University, London, Ontario, Canada
- London Regional Cancer Program, London, Ontario, Canada
- * E-mail:
| | - Maher Kurdi
- Department of Pathology, Western University, London, Ontario, Canada
- Department of Pathology, King Abdulaziz University, Jeddah, Makkah, Saudi Arabia
| | - Yong Wang
- Robarts Research Institute, Western University, London, Ontario, Canada
| | - Baraa Al-Khazraji
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Laura Morrison
- Lawson Imaging, Lawson Health Research Institute, London, Ontario, Canada
| | - Lisa Hoffman
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, Canada
- Lawson Imaging, Lawson Health Research Institute, London, Ontario, Canada
| | - Dwayne Jackson
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Cathie Crukley
- Robarts Research Institute, Western University, London, Ontario, Canada
| | - Ting-Yim Lee
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- Robarts Research Institute, Western University, London, Ontario, Canada
- Lawson Imaging, Lawson Health Research Institute, London, Ontario, Canada
- Department of Medical Imaging, Western University, London, Ontario, Canada
- Department of Oncology, Western University, London, Ontario, Canada
| | - Glenn Bauman
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- Department of Oncology, Western University, London, Ontario, Canada
- London Regional Cancer Program, London, Ontario, Canada
| | - Slav Yartsev
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- Department of Oncology, Western University, London, Ontario, Canada
- London Regional Cancer Program, London, Ontario, Canada
| |
Collapse
|
42
|
Lu J, Zhou J, Cai C, Cai S, Chen Z. Observation of true and pseudo NOE signals using CEST-MRI and CEST-MRS sequences with and without lipid suppression. Magn Reson Med 2014; 73:1615-22. [PMID: 24803172 DOI: 10.1002/mrm.25277] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 04/09/2014] [Accepted: 04/11/2014] [Indexed: 11/08/2022]
Abstract
PURPOSE To investigate the characteristics of nuclear Overhauser enhancement (NOE) imaging signals in the brain at 7T. METHODS Fresh hen eggs, as well as six healthy, and six C6 glioma-bearing Wistar rats were scanned using chemical exchange saturation transfer-magnetic resonance imaging (CEST-MRI) and chemical exchange saturation transfer-magnetic resonance spectroscopy (CEST-MRS) sequences (saturation duration 3 s, power 1.47 µT) with and without lipid suppression. CEST data were acquired over an offset range of -6 to +6 ppm relative to the water resonance in 0.5 ppm steps. RESULTS The water signals were not disrupted by other protons during the CEST-MRS sequences, and true NOE signals could be observed. Using the CEST-MRI sequence without lipid suppression, pseudo NOE imaging signals were observed in the lipid-containing regions (egg yolk, scalp, and even white matter). These pseudo NOE signals were almost (but incompletely) removed with the lipid suppression. Egg yolk results indicated the presence of the NOE to olefinic protons overlapping with the water signal. In vivo experiments showed that the amide proton transfer signal was larger in the tumor, whereas the NOE signal was larger in the normal white matter. CONCLUSIONS True NOE signals can be detected using MRS sequences, and considerable pseudo NOE imaging signals may be observed using MRI sequences.
Collapse
Affiliation(s)
- Jianhua Lu
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China
| | | | | | | | | |
Collapse
|