1
|
Hosoya T, Kambe A, Kesumayadi I, Makishima K, Sueyoshi S, Sakamoto M, Kurosaki M. Mechanism and significance of diffusion restriction followed by calcification in high-grade glioma treated with bevacizumab. Sci Rep 2024; 14:26419. [PMID: 39488647 PMCID: PMC11531518 DOI: 10.1038/s41598-024-78226-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024] Open
Abstract
In this study, we focused on calcification and diffusion restriction, which sometimes appear around the resection cavity or periventricular white matter in patients with high-grade glioma (HGG) treated with bevacizumab (BVZ), as candidate imaging biomarkers for BVZ treatment efficacy. We investigated the timing of the appearance of diffusion restriction and calcification using magnetic resonance imaging and computed tomography in 35 patients with newly diagnosed or recurrent HGG treated with BVZ. In 17 (48.6%) patients, calcification was identified around the resection cavity or periventricular white matter at a median of 12 months after the initiation of BVZ treatment. Patients with calcification had significantly longer progression-free survival (16 vs. 7 months; p = 0.0023) and overall survival (36 vs. 12 months; p = 0.0006) than those without calcification. Histopathological examination revealed the presence of scattered microcalcifications within areas of necrosis, which suggested dystrophic calcification induced by BVZ. Diffusion-restricted lesions that appeared in patients with calcification had significantly lower apparent diffusion coefficients than those in patients without calcifications, indicating the presence of treatment-related necrosis but not hypercellularity. In conclusion, the radiological finding of diffusion restriction followed by calcification could be a potential imaging biomarker for favorable clinical course in patients with HGG treated with BVZ.
Collapse
Affiliation(s)
- Tomohiro Hosoya
- Division of Neurosurgery, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, 36-1, Nishi-cho, Yonago, 683-8504, Tottori, Japan
| | - Atsushi Kambe
- Division of Neurosurgery, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, 36-1, Nishi-cho, Yonago, 683-8504, Tottori, Japan.
| | - Irfan Kesumayadi
- Division of Neurosurgery, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, 36-1, Nishi-cho, Yonago, 683-8504, Tottori, Japan
| | - Karen Makishima
- Division of Pathology, Department of Pathology, Faculty of Medicine, Tottori University, Yonago, 683- 8504, Tottori, Japan
| | - Shuntaro Sueyoshi
- Division of Neurosurgery, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, 36-1, Nishi-cho, Yonago, 683-8504, Tottori, Japan
| | - Makoto Sakamoto
- Division of Neurosurgery, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, 36-1, Nishi-cho, Yonago, 683-8504, Tottori, Japan
| | - Masamichi Kurosaki
- Division of Neurosurgery, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, 36-1, Nishi-cho, Yonago, 683-8504, Tottori, Japan
| |
Collapse
|
2
|
Kambe A, Kitao S, Ochiai R, Hosoya T, Fujii S, Kurosaki M. The utility of arterial spin labeling imaging for predicting prognosis after a recurrence of high-grade glioma in patients under bevacizumab treatment. J Neurooncol 2024; 166:175-183. [PMID: 38165552 DOI: 10.1007/s11060-023-04550-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 12/20/2023] [Indexed: 01/04/2024]
Abstract
BACKGROUND AND PURPOSE Currently, the antiangiogenic agent bevacizumab (BVZ) is used as a treatment option for high-grade glioma (HGG) patients. However, BVZ restores disruptions of the blood-brain barrier, which leads to the disappearance of contrast enhancement during radiological examinations and therefore complicates evaluations of treatment efficacy. This study aimed to investigate the radio-morphological features of recurrent lesions that newly appeared under BVZ therapy, as well as the utility of arterial spin labeling (ASL) perfusion imaging for evaluating treatment response and prognosis in HGG patients receiving BVZ. METHODS Thirty-two patients (20 males, 12 females; age range, 35-84 years) with HGG who experienced a recurrence under BVZ therapy were enrolled. We measured the relative cerebral blood flow (rCBF) values of each recurrent lesion using ASL, and retrospectively investigated the correlation between rCBF values and prognosis. RESULTS The optimal rCBF cut-off value for predicting prognosis was defined as 1.67 using receiver operating characteristic curve analysis. The patients in the rCBF < 1.67 group had significantly longer overall survival (OS) and post-progression survival (PPS) than those in the rCBF ≥ 1.67 group (OS: 34.0 months vs. 13.0 months, p = 0.03 and PPS: 13.0 months vs. 6.0 months, p < 0.001, respectively). CONCLUSION The ASL-derived rCBF values of recurrent lesions may serve as an effective imaging biomarker for prognosis in HGG patients undergoing BVZ therapy. Low rCBF values may indicate that BVZ efficacy is sustainable, which will influence BVZ treatment strategies in HGG patients.
Collapse
Affiliation(s)
- Atsushi Kambe
- Department of Brain and Neurosciences, Division of Neurosurgery, Faculty of Medicine, Tottori University, Tottori, Japan.
| | - Shinichiro Kitao
- Department of Multidisciplinary Internal Medicine, Division of Radiology, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Ryoya Ochiai
- Department of Multidisciplinary Internal Medicine, Division of Radiology, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Tomohiro Hosoya
- Department of Brain and Neurosciences, Division of Neurosurgery, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Shinya Fujii
- Department of Multidisciplinary Internal Medicine, Division of Radiology, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Masamichi Kurosaki
- Department of Brain and Neurosciences, Division of Neurosurgery, Faculty of Medicine, Tottori University, Tottori, Japan
| |
Collapse
|
3
|
Kciuk M, Yahya EB, Mohamed MMI, Abdulsamad MA, Allaq AA, Gielecińska A, Kontek R. Insights into the Role of LncRNAs and miRNAs in Glioma Progression and Their Potential as Novel Therapeutic Targets. Cancers (Basel) 2023; 15:3298. [PMID: 37444408 DOI: 10.3390/cancers15133298] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Accumulating evidence supports that both long non-coding and micro RNAs (lncRNAs and miRNAs) are implicated in glioma tumorigenesis and progression. Poor outcome of gliomas has been linked to late-stage diagnosis and mostly ineffectiveness of conventional treatment due to low knowledge about the early stage of gliomas, which are not possible to observe with conventional diagnostic approaches. The past few years witnessed a revolutionary advance in biotechnology and neuroscience with the understanding of tumor-related molecules, including non-coding RNAs that are involved in the angiogenesis and progression of glioma cells and thus are used as prognostic biomarkers as well as novel therapeutic targets. The emerging research on lncRNAs and miRNAs highlights their crucial role in glioma progression, offering new insights into the disease. These non-coding RNAs hold significant potential as novel therapeutic targets, paving the way for innovative treatment approaches against glioma. This review encompasses a comprehensive discussion about the role of lncRNAs and miRNAs in gene regulation that is responsible for the promotion or the inhibition of glioma progression and collects the existing links between these key cancer-related molecules.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, 90-237 Lodz, Poland
| | - Esam Bashir Yahya
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Penang 11800, Malaysia
| | | | - Muhanad A Abdulsamad
- Department of Molecular Biology, Faculty of Science, Sabratha University, Sabratha 00218, Libya
| | - Abdulmutalib A Allaq
- Faculty of Applied Science, Universiti Teknologi MARA, Shah Alam 40450, Malaysia
| | - Adrianna Gielecińska
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, 90-237 Lodz, Poland
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
| |
Collapse
|
4
|
Ferda J, Frölich M, Ferdová E, Heidenreich F, Charvát R, Mírka H. Neovascularization, vascular mimicry and molecular exchange: The imaging of tumorous tissue aggressiveness based on tissue perfusion. Eur J Radiol 2023; 163:110797. [PMID: 37018901 DOI: 10.1016/j.ejrad.2023.110797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 04/05/2023]
Abstract
Angiogenesis in healthy tissue and within malignant tumors differs on many levels, which may partly be explained by vascular mimicry formation resulting in altered contrast material or different radiopharmaceuticals distributions. Failed remodulation results in changes in the molecular exchange through the capillary wall and those consequences affect the behavior of contrast agents and radiopharmaceuticals. One of the most indicative signs of malignant tissue is the increased permeability and the faster molecular exchange that occurs between the extracellular and intravascular spaces. Dynamic imaging can help to assess the changed microenvironment. The fast-distribution of molecules reflects newly developed conditions in blood-flow redistribution inside a tumor and within the affected organ during the early stages of tumor formation. Tumor development, as well as aggressiveness, can be assessed based on the change to the vascular bed development, the level of molecular exchange within the tissue, and/or indicative distribution within the organ. The study of the vascular network organization and its impact on the distribution of molecules is important to our understanding of the image pattern in several imaging methods, which in turn influences our interpretation of the findings. A hybrid imaging approach (including PET/MRI) allows the quantification of vascularization and/or its pathophysiological impressions in structural and metabolic images. It might optimize the evaluation of the pretreatment imaging, as well as help assess the effect of therapy targeting neovascularization; antiVEGF drugs and embolization-based therapies, for example.
Collapse
Affiliation(s)
- Jiří Ferda
- Department of the Imaging, University Hospital Pilsen and Charles University Medical Faculty in Pilsen, Czech Republic.
| | - Matthias Frölich
- Department of the Imaging, University Hospital Pilsen and Charles University Medical Faculty in Pilsen, Czech Republic; Klinik für Radiologie und Nuklearmedizin, Universitäts Klinikum Mannheim
| | - Eva Ferdová
- Department of the Imaging, University Hospital Pilsen and Charles University Medical Faculty in Pilsen, Czech Republic
| | - Filip Heidenreich
- Department of the Imaging, University Hospital Pilsen and Charles University Medical Faculty in Pilsen, Czech Republic
| | - Radim Charvát
- Department of the Imaging, University Hospital Pilsen and Charles University Medical Faculty in Pilsen, Czech Republic
| | - Hynek Mírka
- Department of the Imaging, University Hospital Pilsen and Charles University Medical Faculty in Pilsen, Czech Republic
| |
Collapse
|
5
|
Barca C, Foray C, Zinnhardt B, Winkeler A, Herrlinger U, Grauer OM, Jacobs AH. In Vivo Quantitative Imaging of Glioma Heterogeneity Employing Positron Emission Tomography. Cancers (Basel) 2022; 14:cancers14133139. [PMID: 35804911 PMCID: PMC9264799 DOI: 10.3390/cancers14133139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma is the most common primary brain tumor, highly aggressive by being proliferative, neovascularized and invasive, heavily infiltrated by immunosuppressive glioma-associated myeloid cells (GAMs), including glioma-associated microglia/macrophages (GAMM) and myeloid-derived suppressor cells (MDSCs). Quantifying GAMs by molecular imaging could support patient selection for GAMs-targeting immunotherapy, drug target engagement and further assessment of clinical response. Magnetic resonance imaging (MRI) and amino acid positron emission tomography (PET) are clinically established imaging methods informing on tumor size, localization and secondary phenomena but remain quite limited in defining tumor heterogeneity, a key feature of glioma resistance mechanisms. The combination of different imaging modalities improved the in vivo characterization of the tumor mass by defining functionally distinct tissues probably linked to tumor regression, progression and infiltration. In-depth image validation on tracer specificity, biological function and quantification is critical for clinical decision making. The current review provides a comprehensive overview of the relevant experimental and clinical data concerning the spatiotemporal relationship between tumor cells and GAMs using PET imaging, with a special interest in the combination of amino acid and translocator protein (TSPO) PET imaging to define heterogeneity and as therapy readouts.
Collapse
Affiliation(s)
- Cristina Barca
- European Institute for Molecular Imaging (EIMI), University of Münster, D-48149 Münster, Germany; (C.F.); (B.Z.)
- Correspondence: (C.B.); (A.H.J.)
| | - Claudia Foray
- European Institute for Molecular Imaging (EIMI), University of Münster, D-48149 Münster, Germany; (C.F.); (B.Z.)
| | - Bastian Zinnhardt
- European Institute for Molecular Imaging (EIMI), University of Münster, D-48149 Münster, Germany; (C.F.); (B.Z.)
- Biomarkers & Translational Technologies (BTT), Pharma Research & Early Development (pRED), F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Alexandra Winkeler
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, F-91401 Orsay, France;
| | - Ulrich Herrlinger
- Division of Clinical Neuro-Oncology, Department of Neurology, University Hospital Bonn, D-53105 Bonn, Germany;
- Centre of Integrated Oncology (CIO), University Hospital Bonn, D-53127 Bonn, Germany
| | - Oliver M. Grauer
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, D-48149 Münster, Germany;
| | - Andreas H. Jacobs
- European Institute for Molecular Imaging (EIMI), University of Münster, D-48149 Münster, Germany; (C.F.); (B.Z.)
- Centre of Integrated Oncology (CIO), University Hospital Bonn, D-53127 Bonn, Germany
- Department of Geriatrics with Neurology, Johanniter Hospital, D-53113 Bonn, Germany
- Correspondence: (C.B.); (A.H.J.)
| |
Collapse
|
6
|
A Comparative and Summative Study of Radiomics-based Overall Survival Prediction in Glioblastoma Patients. J Comput Assist Tomogr 2022; 46:470-479. [PMID: 35405713 DOI: 10.1097/rct.0000000000001300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE This study aimed to assess different machine learning models based on radiomic features, Visually Accessible Rembrandt Images features and clinical characteristics in overall survival prediction of glioblastoma and to identify the reproducible features. MATERIALS AND METHODS Patients with preoperative magnetic resonance scans were allocated into 3 data sets. The Least Absolute Shrinkage and Selection Operator was used for feature selection. The prediction models were built by random survival forest (RSF) and Cox regression. C-index and integrated Brier scores were calculated to compare model performances. RESULTS Patients with cortical involvement had shorter survival times in the training set (P = 0.006). Random survival forest showed higher C-index than Cox, and the RSF model based on the radiomic features was the best one (testing set: C-index = 0.935 ± 0.023). Ten reproducible radiomic features were summarized. CONCLUSIONS The RSF model based on radiomic features had promising potential in predicting overall survival of glioblastoma. Ten reproducible features were identified.
Collapse
|
7
|
Wardak M, Sonni I, Fan AP, Minamimoto R, Jamali M, Hatami N, Zaharchuk G, Fischbein N, Nagpal S, Li G, Koglin N, Berndt M, Bullich S, Stephens AW, Dinkelborg LM, Abel T, Manning HC, Rosenberg J, Chin FT, Sam Gambhir S, Mittra ES. 18F-FSPG PET/CT Imaging of System x C- Transporter Activity in Patients with Primary and Metastatic Brain Tumors. Radiology 2022; 303:620-631. [PMID: 35191738 DOI: 10.1148/radiol.203296] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background The PET tracer (4S)-4-(3-[18F]fluoropropyl)-l-glutamate (18F-FSPG) targets the system xC- cotransporter, which is overexpressed in various tumors. Purpose To assess the role of 18F-FSPG PET/CT in intracranial malignancies. Materials and Methods Twenty-six patients (mean age, 54 years ± 12; 17 men; 48 total lesions) with primary brain tumors (n = 17) or brain metastases (n = 9) were enrolled in this prospective, single-center study (ClinicalTrials.gov identifier: NCT02370563) between November 2014 and March 2016. A 30-minute dynamic brain 18F-FSPG PET/CT scan and a static whole-body (WB) 18F-FSPG PET/CT scan at 60-75 minutes were acquired. Moreover, all participants underwent MRI, and four participants underwent fluorine 18 (18F) fluorodeoxyglucose (FDG) PET imaging. PET parameters and their relative changes were obtained for all lesions. Kinetic modeling was used to estimate the 18F-FSPG tumor rate constants using the dynamic and dynamic plus WB PET data. Imaging parameters were correlated to lesion outcomes, as determined with follow-up MRI and/or pathologic examination. The Mann-Whitney U test or Student t test was used for group mean comparisons. Receiver operating characteristic curve analysis was used for performance comparison of different decision measures. Results 18F-FSPG PET/CT helped identify all 48 brain lesions. The mean tumor-to-background ratio (TBR) on the whole-brain PET images at the WB time point was 26.6 ± 24.9 (range: 2.6-150.3). When 18F-FDG PET was performed, 18F-FSPG permitted visualization of non-18F-FDG-avid lesions or allowed better lesion differentiation from surrounding tissues. In participants with primary brain tumors, the predictive accuracy of the relative changes in influx rate constant Ki and maximum standardized uptake value to discriminate between poor and good lesion outcomes were 89% and 81%, respectively. There were significant differences in the 18F-FSPG uptake curves of lesions with good versus poor outcomes in the primary brain tumor group (P < .05) but not in the brain metastases group. Conclusion PET/CT imaging with (4S)-4-(3-[18F]fluoropropyl)-l-glutamate (18F-FSPG) helped detect primary brain tumors and brain metastases with a high tumor-to-background ratio. Relative changes in 18F-FSPG uptake with multi-time-point PET appear to be helpful in predicting lesion outcomes. Clinical trial registration no. NCT02370563 © RSNA, 2022 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Mirwais Wardak
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Ida Sonni
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Audrey P Fan
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Ryogo Minamimoto
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Mehran Jamali
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Negin Hatami
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Greg Zaharchuk
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Nancy Fischbein
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Seema Nagpal
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Gordon Li
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Norman Koglin
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Mathias Berndt
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Santiago Bullich
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Andrew W Stephens
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Ludger M Dinkelborg
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Ty Abel
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - H Charles Manning
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Jarrett Rosenberg
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Frederick T Chin
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Sanjiv Sam Gambhir
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Erik S Mittra
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| |
Collapse
|
8
|
Chiaravalloti A, Cimini A, Ricci M, Quartuccio N, Arnone G, Filippi L, Calabria F, Leporace M, Bagnato A, Schillaci O. Positron emission tomography imaging in primary brain tumors. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00042-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
9
|
Yao H, Liu J, Zhang C, Shao Y, Li X, Yu Z, Huang Y. Apatinib inhibits glioma cell malignancy in patient-derived orthotopic xenograft mouse model by targeting thrombospondin 1/myosin heavy chain 9 axis. Cell Death Dis 2021; 12:927. [PMID: 34635636 PMCID: PMC8505401 DOI: 10.1038/s41419-021-04225-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/09/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022]
Abstract
We determined the antitumor mechanism of apatinib in glioma using a patient-derived orthotopic xenograft (PDOX) glioma mouse model and glioblastoma (GBM) cell lines. The PDOX mouse model was established using tumor tissues from two glioma patients via single-cell injections. Sixteen mice were successfully modeled and randomly divided into two equal groups (n = 8/group): apatinib and normal control. Survival analysis and in vivo imaging was performed to determine the effect of apatinib on glioma proliferation in vivo. Candidate genes in GBM cells that may be affected by apatinib treatment were screened using RNA-sequencing coupled with quantitative mass spectrometry, data mining of The Cancer Genome Atlas, and Chinese Glioma Genome Atlas databases, and immunohistochemistry analysis of clinical high-grade glioma pathology samples. Quantitative reverse transcription-polymerase chain reaction (qPCR), western blotting, and co-immunoprecipitation (co-IP) were performed to assess gene expression and the apatinib-mediated effect on glioma cell malignancy. Apatinib inhibited the proliferation and malignancy of glioma cells in vivo and in vitro. Thrombospondin 1 (THBS1) was identified as a potential target of apatinib that lead to inhibited glioma cell proliferation. Apatinib-mediated THBS1 downregulation in glioma cells was confirmed by qPCR and western blotting. Co-IP and mass spectrometry analysis revealed that THBS1 could interact with myosin heavy chain 9 (MYH9) in glioma cells. Simultaneous THBS1 overexpression and MYH9 knockdown suppressed glioma cell invasion and migration. These data suggest that apatinib targets THBS1 in glioma cells, potentially via MYH9, to inhibit glioma cell malignancy and may provide novel targets for glioma therapy.
Collapse
Affiliation(s)
- Hui Yao
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, No188, Shizi Street, Suzhou, 215007, Jiangsu, China
| | - Jiangang Liu
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, No188, Shizi Street, Suzhou, 215007, Jiangsu, China
| | - Chi Zhang
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, No188, Shizi Street, Suzhou, 215007, Jiangsu, China
| | - Yunxiang Shao
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, No188, Shizi Street, Suzhou, 215007, Jiangsu, China
| | - Xuetao Li
- Department of Neurosurgery, Dushu Lake Hospital Affiliated of Soochow University, Suzhou, 215124, Jiangsu, China
| | - Zhengquan Yu
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, No188, Shizi Street, Suzhou, 215007, Jiangsu, China.
| | - Yulun Huang
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, No188, Shizi Street, Suzhou, 215007, Jiangsu, China.
- Department of Neurosurgery, Dushu Lake Hospital Affiliated of Soochow University, Suzhou, 215124, Jiangsu, China.
| |
Collapse
|
10
|
Fully automated analysis combining [ 18F]-FET-PET and multiparametric MRI including DSC perfusion and APTw imaging: a promising tool for objective evaluation of glioma progression. Eur J Nucl Med Mol Imaging 2021; 48:4445-4455. [PMID: 34173008 PMCID: PMC8566389 DOI: 10.1007/s00259-021-05427-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/24/2021] [Indexed: 12/15/2022]
Abstract
Purpose To evaluate diagnostic accuracy of fully automated analysis of multimodal imaging data using [18F]-FET-PET and MRI (including amide proton transfer-weighted (APTw) imaging and dynamic-susceptibility-contrast (DSC) perfusion) in differentiation of tumor progression from treatment-related changes in patients with glioma. Material and methods At suspected tumor progression, MRI and [18F]-FET-PET data as part of a retrospective analysis of an observational cohort of 66 patients/74 scans (51 glioblastoma and 23 lower-grade-glioma, 8 patients included at two different time points) were automatically segmented into necrosis, FLAIR-hyperintense, and contrast-enhancing areas using an ensemble of deep learning algorithms. In parallel, previous MR exam was processed in a similar way to subtract preexisting tumor areas and focus on progressive tumor only. Within these progressive areas, intensity statistics were automatically extracted from [18F]-FET-PET, APTw, and DSC-derived cerebral-blood-volume (CBV) maps and used to train a Random Forest classifier with threefold cross-validation. To evaluate contribution of the imaging modalities to the classifier’s performance, impurity-based importance measures were collected. Classifier performance was compared with radiology reports and interdisciplinary tumor board assessments. Results In 57/74 cases (77%), tumor progression was confirmed histopathologically (39 cases) or via follow-up imaging (18 cases), while remaining 17 cases were diagnosed as treatment-related changes. The classification accuracy of the Random Forest classifier was 0.86, 95% CI 0.77–0.93 (sensitivity 0.91, 95% CI 0.81–0.97; specificity 0.71, 95% CI 0.44–0.9), significantly above the no-information rate of 0.77 (p = 0.03), and higher compared to an accuracy of 0.82 for MRI (95% CI 0.72–0.9), 0.81 for [18F]-FET-PET (95% CI 0.7–0.89), and 0.81 for expert consensus (95% CI 0.7–0.89), although these differences were not statistically significant (p > 0.1 for all comparisons, McNemar test). [18F]-FET-PET hot-spot volume was single-most important variable, with relevant contribution from all imaging modalities. Conclusion Automated, joint image analysis of [18F]-FET-PET and advanced MR imaging techniques APTw and DSC perfusion is a promising tool for objective response assessment in gliomas. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-021-05427-8.
Collapse
|
11
|
Bliesener Y, Lebel RM, Acharya J, Frayne R, Nayak KS. Pseudo Test-Retest Evaluation of Millimeter-Resolution Whole-Brain Dynamic Contrast-enhanced MRI in Patients with High-Grade Glioma. Radiology 2021; 300:410-420. [PMID: 34100683 PMCID: PMC8328086 DOI: 10.1148/radiol.2021203628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Background Advances in sub-Nyquist–sampled dynamic contrast-enhanced (DCE) MRI enable monitoring of brain tumors with millimeter resolution and whole-brain coverage. Such undersampled quantitative methods need careful characterization regarding achievable test-retest reproducibility. Purpose To demonstrate a fully automated high-resolution whole-brain DCE MRI pipeline with 30-fold sparse undersampling and estimate its reproducibility on the basis of reference regions of stable tissue types during multiple posttreatment time points by using longitudinal clinical images of high-grade glioma. Materials and Methods Two methods for sub-Nyquist–sampled DCE MRI were extended with automatic estimation of vascular input functions. Continuously acquired three-dimensional k-space data with ramped-up flip angles were partitioned to yield high-resolution, whole-brain tracer kinetic parameter maps with matched precontrast-agent T1 and M0 maps. Reproducibility was estimated in a retrospective study in participants with high-grade glioma, who underwent three consecutive standard-of-care examinations between December 2016 and April 2019. Coefficients of variation and reproducibility coefficients were reported for histogram statistics of the tracer kinetic parameters plasma volume fraction and volume transfer constant (Ktrans) on five healthy tissue types. Results The images from 13 participants (mean age ± standard deviation, 61 years ± 10; nine women) with high-grade glioma were evaluated. In healthy tissues, the protocol achieved a coefficient of variation less than 57% for median Ktrans, if Ktrans was estimated consecutively. The maximum reproducibility coefficient for median Ktrans was estimated to be at 0.06 min–1 for large or low-enhancing tissues and to be as high as 0.48 min–1 in smaller or strongly enhancing tissues. Conclusion A fully automated, sparsely sampled DCE MRI reconstruction with patient-specific vascular input function offered high spatial and temporal resolution and whole-brain coverage; in healthy tissues, the protocol estimated median volume transfer constant with maximum reproducibility coefficient of 0.06 min–1 in large, low-enhancing tissue regions and maximum reproducibility coefficient of less than 0.48 min–1 in smaller or more strongly enhancing tissue regions. Published under a CC BY 4.0 license. Online supplemental material is available for this article. See also the editorial by Lenkinski in this issue.
Collapse
Affiliation(s)
- Yannick Bliesener
- From the Ming Hsieh Department of Electrical and Computer Engineering, Viterbi School of Engineering, University of Southern California, 3740 McClintock Ave, EEB 400, Los Angeles, CA 90089-2564 (Y.B., K.S.N.); GE Healthcare, Calgary, Canada (R.M.L.); Department of Radiology, University of Calgary, Calgary, Canada (R.M.L.); Seaman Family MR Research Centre, Foothills Hospital, Calgary, Canada (R.M.L., R.F.); Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, Calif (J.A., K.S.N.); and Departments of Radiology and Clinical Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.F.)
| | - R Marc Lebel
- From the Ming Hsieh Department of Electrical and Computer Engineering, Viterbi School of Engineering, University of Southern California, 3740 McClintock Ave, EEB 400, Los Angeles, CA 90089-2564 (Y.B., K.S.N.); GE Healthcare, Calgary, Canada (R.M.L.); Department of Radiology, University of Calgary, Calgary, Canada (R.M.L.); Seaman Family MR Research Centre, Foothills Hospital, Calgary, Canada (R.M.L., R.F.); Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, Calif (J.A., K.S.N.); and Departments of Radiology and Clinical Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.F.)
| | - Jay Acharya
- From the Ming Hsieh Department of Electrical and Computer Engineering, Viterbi School of Engineering, University of Southern California, 3740 McClintock Ave, EEB 400, Los Angeles, CA 90089-2564 (Y.B., K.S.N.); GE Healthcare, Calgary, Canada (R.M.L.); Department of Radiology, University of Calgary, Calgary, Canada (R.M.L.); Seaman Family MR Research Centre, Foothills Hospital, Calgary, Canada (R.M.L., R.F.); Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, Calif (J.A., K.S.N.); and Departments of Radiology and Clinical Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.F.)
| | - Richard Frayne
- From the Ming Hsieh Department of Electrical and Computer Engineering, Viterbi School of Engineering, University of Southern California, 3740 McClintock Ave, EEB 400, Los Angeles, CA 90089-2564 (Y.B., K.S.N.); GE Healthcare, Calgary, Canada (R.M.L.); Department of Radiology, University of Calgary, Calgary, Canada (R.M.L.); Seaman Family MR Research Centre, Foothills Hospital, Calgary, Canada (R.M.L., R.F.); Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, Calif (J.A., K.S.N.); and Departments of Radiology and Clinical Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.F.)
| | - Krishna S Nayak
- From the Ming Hsieh Department of Electrical and Computer Engineering, Viterbi School of Engineering, University of Southern California, 3740 McClintock Ave, EEB 400, Los Angeles, CA 90089-2564 (Y.B., K.S.N.); GE Healthcare, Calgary, Canada (R.M.L.); Department of Radiology, University of Calgary, Calgary, Canada (R.M.L.); Seaman Family MR Research Centre, Foothills Hospital, Calgary, Canada (R.M.L., R.F.); Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, Calif (J.A., K.S.N.); and Departments of Radiology and Clinical Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.F.)
| |
Collapse
|
12
|
Solnes LB, Jacobs AH, Coughlin JM, Du Y, Goel R, Hammoud DA, Pomper MG. Central Nervous System Molecular Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00088-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
13
|
Li D, Patel CB, Xu G, Iagaru A, Zhu Z, Zhang L, Cheng Z. Visualization of Diagnostic and Therapeutic Targets in Glioma With Molecular Imaging. Front Immunol 2020; 11:592389. [PMID: 33193439 PMCID: PMC7662122 DOI: 10.3389/fimmu.2020.592389] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/08/2020] [Indexed: 02/04/2023] Open
Abstract
Gliomas, particularly high-grade gliomas including glioblastoma (GBM), represent the most common and malignant types of primary brain cancer in adults, and carry a poor prognosis. GBM has been classified into distinct subgroups over the years based on cellular morphology, clinical characteristics, biomarkers, and neuroimaging findings. Based on these classifications, differences in therapeutic response and patient outcomes have been established. Recently, the identification of complex molecular signatures of GBM has led to the development of diverse targeted therapeutic regimens and translation into multiple clinical trials. Chemical-, peptide-, antibody-, and nanoparticle-based probes have been designed to target specific molecules in gliomas and then be visualized with multimodality molecular imaging (MI) techniques including positron emission tomography (PET), single-photon emission computed tomography (SPECT), near-infrared fluorescence (NIRF), bioluminescence imaging (BLI), and magnetic resonance imaging (MRI). Thus, multiple molecules of interest can now be noninvasively imaged to guide targeted therapies with a potential survival benefit. Here, we review developments in molecular-targeted diagnosis and therapy in glioma, MI of these targets, and MI monitoring of treatment response, with a focus on the biological mechanisms of these advanced molecular probes. MI probes have the potential to noninvasively demonstrate the pathophysiologic features of glioma for diagnostic, treatment, and response assessment considerations for various targeted therapies, including immunotherapy. However, most MI tracers are in preclinical development, with only integrin αVβ3 and isocitrate dehydrogenase (IDH)-mutant MI tracers having been translated to patients. Expanded international collaborations would accelerate translational research in the field of glioma MI.
Collapse
Affiliation(s)
- Deling Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China National Clinical Research Center for Neurological Diseases (NCRC-ND), Beijing, China
| | - Chirag B. Patel
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, United States
- Division of Neuro-Oncology, Department of Neurology and Neurological Sciences, School of Medicine, Stanford University, Stanford, CA, United States
| | - Guofan Xu
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Andrei Iagaru
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Zhaohui Zhu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China National Clinical Research Center for Neurological Diseases (NCRC-ND), Beijing, China
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
14
|
Xue W, Ton H, Zhang J, Xie T, Chen X, Zhou B, Guo Y, Fang J, Wang S, Zhang W. Patient‑derived orthotopic xenograft glioma models fail to replicate the magnetic resonance imaging features of the original patient tumor. Oncol Rep 2020; 43:1619-1629. [PMID: 32323818 PMCID: PMC7107810 DOI: 10.3892/or.2020.7538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/12/2020] [Indexed: 12/14/2022] Open
Abstract
Patient-derived orthotopic glioma xenograft models are important platforms used for pre-clinical research of glioma. In the present study, the diagnostic ability of magnetic resonance imaging (MRI) was examined with regard to the identification of biomarkers obtained from patient-derived glioma xenografts and human tumors. Conventional MRI, diffusion weighted imaging and dynamic contrast-enhanced (DCE)-MRI were used to analyze seven pairs of high grade gliomas with their corresponding xenografts obtained from non-obese diabetic-severe-combined immunodeficiency nude mice. Tumor samples were collected for transcriptome sequencing and histopathological staining, and differentially expressed genes were screened between the original tumors and the corresponding xenografts. Gene Ontology (GO) analysis was performed to predict the functions of these genes. In 6 cases of xenografts with diffuse growth, the degree of enhancement was significantly lower compared with the original tumors. Histopathological staining indicated that the microvascular area and microvascular diameter of the xenografts were significantly lower compared with the original tumors (P=0.009 and P=0.007, respectively). In one case, there was evidence of nodular tumor growth in the mouse. Both MRI and histopathological staining showed a clear demarcation between the transplanted tumors and the normal brain tissues. The relative apparent diffusion coefficient values of the 7 cases examined were significantly higher compared with the corresponding original tumors (P=0.001) and transfer coefficient values derived from DCE-MRI of the tumor area was significantly lower compared with the original tumors (P=0.016). GO analysis indicated that the expression levels of extracellular matrix-associated genes, angiogenesis-associated genes and immune function-associated genes in the original tumors were higher compared with the corresponding xenografts. In conclusion, the data demonstrated that the MRI features of patient-derived xenograft glioma models in mice were different compared with those of the original patient tumors. Differential gene expression may underlie the differences noted in the MRI features between original tumors and corresponding xenografts. The results of the present study highlight the precautions that should be taken when extrapolating data from patient-derived xenograft studies, and their applicability to humans.
Collapse
Affiliation(s)
- Wei Xue
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Haipeng Ton
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Junfeng Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Tian Xie
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Xiao Chen
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Bo Zhou
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Yu Guo
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Jingqin Fang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Shunan Wang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| | - Weiguo Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China
| |
Collapse
|
15
|
Chang Y, Donglan Y, Xinchong S, Ganhua L, Bing Z, Yao L, Rutong Z, Qiao H, Xiangsong Z. One-day protocol for 18F-FDG and 13N-ammonia PET/CT with uptake decoupling score in differentiating untreated low-grade glioma from inflammation. Rev Esp Med Nucl Imagen Mol 2020; 39:68-74. [PMID: 32005511 DOI: 10.1016/j.remn.2019.08.001] [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: 04/18/2019] [Revised: 07/27/2019] [Accepted: 08/27/2019] [Indexed: 10/25/2022]
Abstract
PURPOSE Accurate identification of low-grade gliomas (LGGs; World Health Organization grades I and II) and their differentiation from brain inflammation lesions (BILs) remains difficult; however, it is essential for treatment. This study assessed whether a one-day protocol for voxel-wise 18F-FDG and 13N-ammonia PET/CT with uptake decoupling analysis could differentiate LGGs from BILs. MATERIALS AND METHODS Twenty-eight patients with LGGs and 16 patients with BILs underwent 18F-FDG and 13N-ammonia PET/CT on the same day before any type of therapy. The decoupling score and tumor-to-normal tissue (T/N) ratio of 18F-FDG and 13N-ammonia were calculated at each location. Student's t-test was used to compare values, and ROC curve analysis was used to establish a cut-off value for the T/N ratio and decoupling score. Area under the curve (AUC) was calculated to evaluate differential efficacy. RESULTS Significant differences were observed in 13N-ammonia T/N ratio (p=0.018) and decoupling score (p=0.003) between LGGs and BILs; however, the 18F-FDG T/N ratio did not show any differences (p=0.413). Optimal cut-off values for 18F-FDG T/N ratio, 13N-ammonia T/N ratio, and decoupling score were 0.73, 0.97, and 2.31, respectively, with corresponding AUCs of 0.48, 0.68, and 0.77. The respective sensitivity, specificity, and accuracy parameters using these cut-off values were 53.6%, 62.5%, and 56.8%, respectively, for 18F-FDG; 50.0%, 75.0%, and 59.1%, respectively, for 13N-ammonia; and 60.7%, 93.8%, and 72.7%, respectively, for decoupling score. CONCLUSIONS 18F-FDG/13N-ammonia uptake decoupling score can be used to discriminate between LGGs and BILs. Use of a decoupling map of these two tracers can improve visual analysis and diagnostic accuracy.
Collapse
Affiliation(s)
- Y Chang
- Department of Nuclear Medicine, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Y Donglan
- Department of Medical Engineering, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - S Xinchong
- Department of Nuclear Medicine, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - L Ganhua
- Department of Nuclear Medicine, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Z Bing
- Department of Nuclear Medicine, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - L Yao
- School of Data and Computer Science, Sun Yat-Sen University, Guangzhou, China
| | - Z Rutong
- School of Data and Computer Science, Sun Yat-Sen University, Guangzhou, China
| | - H Qiao
- Department of Nuclear Medicine, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Z Xiangsong
- Department of Nuclear Medicine, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
16
|
Graham MS, Krebs S, Bale T, Domfe K, Lobaugh SM, Zhang Z, Dunphy MP, Kaley T, Young RJ. Value of [ 18F]-FDG positron emission tomography in patients with recurrent glioblastoma receiving bevacizumab. Neurooncol Adv 2020; 2:vdaa050. [PMID: 32642703 PMCID: PMC7236386 DOI: 10.1093/noajnl/vdaa050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Treatment of recurrent glioblastoma (GBM) with bevacizumab can induce MRI changes that confound the determination of progression. We sought to determine the value of [18F]-fluorodeoxyglucose (FDG) positron emission tomography (PET) in GBM patients receiving bevacizumab at the time of suspected progression and, thereby, its utility as a potential prognostic adjunct in progressive disease. METHODS This retrospective study included patients who underwent brain FDG PET within 4 weeks of receiving bevacizumab for recurrent GBM with suspected progression. Volumes-of-interest were placed over the reference lesion with measurement of maximum standardized uptake value (SUVmax), peak standardized uptake value (SUVpeak), metabolic tumor volume, total lesion glycolysis (TLG), and tumor-to-normal contralateral white matter ratios (TNR-WM). Tumors were additionally categorized as non-avid or avid based on qualitative FDG uptake. Associations between baseline variables and overall survival (OS) were examined using univariable and multivariable Cox proportional hazards regression, with P < .05 considered significant. RESULTS Thirty-one patients were analyzed. Qualitative FDG uptake was significantly associated with OS (P = .03), with a median OS of 9.0 months in non-avid patients versus 4.5 months in avid patients. SUVmax, SUVpeak, TNR-WM, and TLG were significantly associated with OS (P < .001, TLG: P = .009). FDG avidity and SUVmax remained significantly associated with OS (P = .046 and .048, respectively) in the multivariable analysis including age, KPS, and MGMT status. Dichotomizing patients using an SUVmax cutoff of 15.3 was associated with OS (adjusted P = .048). CONCLUSION FDG PET is a promising imaging tool to further stratify prognosis in recurrent GBM patients on antiangiogenic therapy.
Collapse
Affiliation(s)
- Maya S Graham
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- The Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Simone Krebs
- Department of Radiology, Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Tejus Bale
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kwaku Domfe
- College of Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Stephanie M Lobaugh
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Zhigang Zhang
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mark P Dunphy
- Department of Radiology, Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Thomas Kaley
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- The Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Robert J Young
- Department of Radiology, Neuroradiology Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- The Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
17
|
Chiaravalloti A, Filippi L, Ricci M, Cimini A, Schillaci O. Molecular Imaging in Pediatric Brain Tumors. Cancers (Basel) 2019; 11:cancers11121853. [PMID: 31771237 PMCID: PMC6966547 DOI: 10.3390/cancers11121853] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/16/2019] [Accepted: 11/19/2019] [Indexed: 02/07/2023] Open
Abstract
In the last decade, several radiopharmaceuticals have been developed and investigated for imaging in vivo of pediatric brain tumors with the aim of exploring peculiar metabolic processes as glucose consumption, amino-acid metabolism, and protein synthesis with nuclear medicine techniques. Although the clinical shreds of evidence are limited, preliminary results are encouraging. In this review, we performed web-based and desktop research summarizing the most relevant findings of the literature published to date on this topic. Particular attention was given to the wide spectrum of nuclear medicine advances and trends in pediatric neurooncology and neurosurgery. Furthermore, the role of somatostatin receptor imaging through single-photon emission computed tomography (SPECT) and positron emission tomography (PET) probes, with reference to their potential therapeutic implications, was examined in the peculiar context. Preliminary results show that functional imaging in pediatric brain tumors might lead to significant improvements in terms of diagnostic accuracy and it could be of help in the management of the disease.
Collapse
Affiliation(s)
- Agostino Chiaravalloti
- Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy (O.S.)
- Nuclear Medicine Section, IRCCS Neuromed, 86077 Pozzilli, Italy
- Correspondence: or ; Tel.: +39-062-090-2457
| | - Luca Filippi
- Nuclear Medicine Section, “Santa Maria Goretti” Hospital, 04100 Latina, Italy;
| | - Maria Ricci
- Department of Radiological, Oncological and Pathological Sciences, Faculty of Medicine and Surgery, La Sapienza University, 00161 Rome, Italy;
| | - Andrea Cimini
- Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy (O.S.)
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy (O.S.)
- Nuclear Medicine Section, IRCCS Neuromed, 86077 Pozzilli, Italy
| |
Collapse
|
18
|
Zeiner PS, Kinzig M, Divé I, Maurer GD, Filipski K, Harter PN, Senft C, Bähr O, Hattingen E, Steinbach JP, Sörgel F, Voss M, Steidl E, Ronellenfitsch MW. Regorafenib CSF Penetration, Efficacy, and MRI Patterns in Recurrent Malignant Glioma Patients. J Clin Med 2019; 8:jcm8122031. [PMID: 31766326 PMCID: PMC6947028 DOI: 10.3390/jcm8122031] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/28/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022] Open
Abstract
(1) Background: The phase 2 Regorafenib in Relapsed Glioblastoma (REGOMA) trial indicated a survival benefit for patients with first recurrence of a glioblastoma when treated with the multikinase inhibitor regorafenib (REG) instead of lomustine. The aim of this retrospective study was to investigate REG penetration to cerebrospinal fluid (CSF), treatment efficacy, and effects on magnetic resonance imaging (MRI) in patients with recurrent high-grade gliomas. (2) Methods: Patients were characterized by histology, adverse events, steroid treatment, overall survival (OS), and MRI growth pattern. REG and its two active metabolites were quantified by liquid chromatography/tandem mass spectrometry in patients’ serum and CSF. (3) Results: 21 patients mainly with IDH-wildtype glioblastomas who had been treated with REG were retrospectively identified. Thirteen CFS samples collected from 3 patients of the cohort were available for pharmacokinetic testing. CSF levels of REG and its metabolites were significantly lower than in serum. Follow-up MRI was available in 19 patients and showed progressive disease (PD) in all but 2 patients. Two distinct MRI patterns were identified: 7 patients showed classic PD with progression of contrast enhancing lesions, whereas 11 patients showed a T2-dominant MRI pattern characterized by a marked reduction of contrast enhancement. Median OS was significantly better in patients with a T2-dominant growth pattern (10 vs. 27 weeks respectively, p = 0.003). Diffusion restrictions were observed in 13 patients. (4) Conclusion: REG and its metabolites were detectable in CSF. A distinct MRI pattern that might be associated with an improved OS was observed in half of the patient cohort. Treatment response in the total cohort was poor.
Collapse
Affiliation(s)
- Pia S. Zeiner
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - Martina Kinzig
- IBMP—Institute for Biomedical and Pharmaceutical Research, 90562 Nürnberg-Heroldsberg, Germany; (M.K.); (F.S.)
| | - Iris Divé
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - Gabriele D. Maurer
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
| | - Katharina Filipski
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Institute of Neurology (Edinger-Institute), University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany
| | - Patrick N. Harter
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
- Institute of Neurology (Edinger-Institute), University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany
| | - Christian Senft
- Department of Neurosurgery, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany;
| | - Oliver Bähr
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- Department of Neurology, Klinikum Aschaffenburg-Alzenau, 63739 Aschaffenburg, Germany
| | - Elke Hattingen
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Department of Neuroradiology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany
| | - Joachim P. Steinbach
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - Fritz Sörgel
- IBMP—Institute for Biomedical and Pharmaceutical Research, 90562 Nürnberg-Heroldsberg, Germany; (M.K.); (F.S.)
- Institute of Pharmacology, University Duisburg-Essen, 45141 Essen, Germany
| | - Martin Voss
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - Eike Steidl
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Department of Neuroradiology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany
| | - Michael W. Ronellenfitsch
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
- Correspondence: ; Tel.: +49-69-6301-87711; Fax: +49-69-6301-87713
| |
Collapse
|
19
|
Comparisons Between PET With 11C-Methyl-L-Methionine and Arterial Spin Labeling Perfusion Imaging in Recurrent Glioblastomas Treated With Bevacizumab. Clin Nucl Med 2019; 44:186-193. [PMID: 30562194 DOI: 10.1097/rlu.0000000000002417] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE The aim of this study was to clarify whether arterial spin labeling (ASL) perfusion imaging can assess biological effects from bevacizumab (BEV) therapy as reliably as PET with C-methyl-L-methionine (C-met-PET). MATERIALS AND METHODS Twenty-four patients with recurrent glioblastoma were examined using both ASL and C-met-PET before and 4 and 8 weeks after starting BEV treatment. Tumor-to-normal brain (T/N) ratios, fluctuations in T/N ratio, and tumor volumes were compared between ASL and C-met-PET. Accuracy of predicting patient with long progression-free survival (PFS) was assessed for T/N ratios and fluctuations for ASL and C-met-PET in each phase and in each period using receiver operating characteristic curves. Between 2 groups of patients assigned by cutoff values from receiver operating characteristic curves, PFS was compared in each phase or in each period. RESULTS T/N ratios, fluctuations in ratio, and tumor volumes correlated significantly between ASL and C-met-PET at all time points and all periods. Arterial spin labeling was eligible as a predictor for long PFS only in assessment of fluctuations in T/N ratio. However, the most accurate predictors for long PFS were T/N ratio from C-met-PET at 8 weeks and the fluctuation from baseline to 4 weeks in T/N ratio from C-met-PET. CONCLUSIONS Blood flows on ASL correlated with accumulations of C-met on PET in recurrent glioblastoma under BEV treatment. Although C-met-PET offered superior accuracy for predicting patients with long PFS from time points, ASL offered reliable prediction of long PFS, provided that fluctuations in T/N ratio between consecutive scans are assessed.
Collapse
|
20
|
Shah NJ, da Silva NA, Yun SD. Perfusion weighted imaging using combined gradient/spin echo EPIK: Brain tumour applications in hybrid MR-PET. Hum Brain Mapp 2019; 42:4144-4154. [PMID: 30761676 DOI: 10.1002/hbm.24537] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 01/23/2019] [Accepted: 01/25/2019] [Indexed: 01/30/2023] Open
Abstract
Advanced perfusion-weighted imaging (PWI) methods that combine gradient echo (GE) and spin echo (SE) data are important tools for the study of brain tumours. In PWI, single-shot, EPI-based methods have been widely used due to their relatively high imaging speed. However, when used with increasing spatial resolution, single-shot EPI methods often show limitations in whole-brain coverage for multi-contrast applications. To overcome this limitation, this work employs a new version of EPI with keyhole (EPIK) to provide five echoes: two with GEs, two with mixed GESE and one with SE; the sequence is termed "GESE-EPIK." The performance of GESE-EPIK is evaluated against its nearest relative, EPI, in terms of the temporal signal-to-noise ratio (tSNR). Here, data from brain tumour patients were acquired using a hybrid 3T MR-BrainPET scanner. GESE-EPIK resulted in reduced susceptibility artefacts, shorter TEs for the five echoes and increased brain coverage when compared to EPI. Moreover, compared to EPI, EPIK achieved a comparable tSNR for the first and second echoes and significantly higher tSNR for other echoes. A new method to obtain multi-echo GE and SE data with shorter TEs and increased brain coverage is demonstrated. As proposed here, the workflow can be shortened and the integration of multimodal clinical MR-PET studies can be facilitated.
Collapse
Affiliation(s)
- N Jon Shah
- Institute of Neuroscience and Medicine - 4, Medical Imaging Physics, Forschungszentrum Jülich GmbH, Jülich, Germany.,Institute of Neuroscience-11, Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH, Jülich, Germany.,Department of Neurology, Faculty of Medicine, JARA, RWTH Aachen University, Aachen, Germany.,Monash Biomedical Imaging, School of Psychological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Nuno André da Silva
- Institute of Neuroscience and Medicine - 4, Medical Imaging Physics, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Seong Dae Yun
- Institute of Neuroscience and Medicine - 4, Medical Imaging Physics, Forschungszentrum Jülich GmbH, Jülich, Germany
| |
Collapse
|
21
|
The role of 13N-ammonia in the differential diagnosis of gliomas and brain inflammatory lesions. Ann Nucl Med 2018; 33:61-67. [PMID: 30350180 DOI: 10.1007/s12149-018-1308-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 09/30/2018] [Indexed: 10/28/2022]
Abstract
OBJECTIVE To investigate the utility of 13N-ammonia PET/CT imaging in the differential diagnosis of gliomas and brain inflammations. METHODS 13N-ammonia PET/CT imaging data of 77 patients with gliomas and 34 patients with brain inflammations were retrospectively analyzed. No patients received any treatment before 13N-ammonia imaging. All the patients were diagnosed by stereotactic biopsy or clinical follow-up. Visual and semi-quantitative analysis was performed to analyze the results of 13N-ammonia imaging. Finally, the uptake ratios of each lesion were calculated and its differences among different groups were tested with one-way ANOVA. RESULTS 29.4% inflammations, 51.6% low-grade gliomas and 91.3% high-grade gliomas were positive by visual analysis in 13N-ammonia imaging. The sensitivity, specificity and accuracy for the diagnosis of gliomas were 75.3%, 55.8% and 67.8%, respectively. As for semi-quantitative analysis, the T/G ratios of inflammatory lesions, low-grade gliomas and high-grade gliomas were 0.88 ± 0.24, 1.04 ± 0.43 and 1.43 ± 0.49, respectively. One-way ANOVA revealed that the T/G ratios of high-grade gliomas were significantly higher than those of low-grade gliomas and inflammations (P < 0.05), but there was no statistical difference between low-grade gliomas and inflammations (P = 0.118). Among the inflammatory lesions, T/G ratios were not statistically different between infectious and demyelinating lesions (P > 0.05). ROC curve analysis showed that the optimal cut-off value of T/G ratio in distinguishing gliomas from inflammations was 1.21 with the AUC 0.78. The sensitivity, specificity, accuracy, PPV and NPV were 52.9%, 94.4%, 65.3%, 95.7% and 45.9%, respectively. ROC curve analysis showed that the optimal cut-off value of T/G ratio in distinguishing high-grade gliomas from low-grade gliomas was 1.06 with the AUC 0.78. The sensitivity, specificity, accuracy, PPV and NPV were 81.5%, 67.7%, 76.5%, 81.5% and 67.7%, respectively. ROC curve analysis showed that the optimal cut-off value of T/G ratio in distinguishing high-grade gliomas from low-grade gliomas and inflammations was 1.19 with the AUC 0.84. The sensitivity, specificity, accuracy, PPV and NPV were 70.4%, 85.1%, 78.5%, 79.2% and 78.1%, respectively. CONCLUSIONS 13N-ammonia imaging is effective in distinguishing high-grade gliomas from low-grade gliomas and inflammations, but its role in the differential diagnosis of low-grade gliomas and brain inflammatory lesions is limited, and the accuracy needs to be improved.
Collapse
|
22
|
Serial FLT PET imaging to discriminate between true progression and pseudoprogression in patients with newly diagnosed glioblastoma: a long-term follow-up study. Eur J Nucl Med Mol Imaging 2018; 45:2404-2412. [PMID: 30032322 PMCID: PMC6208814 DOI: 10.1007/s00259-018-4090-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/09/2018] [Indexed: 12/23/2022]
Abstract
Purpose Response evaluation in patients with glioblastoma after chemoradiotherapy is challenging due to progressive, contrast-enhancing lesions on MRI that do not reflect true tumour progression. In this study, we prospectively evaluated the ability of the PET tracer 18F-fluorothymidine (FLT), a tracer reflecting proliferative activity, to discriminate between true progression and pseudoprogression in newly diagnosed glioblastoma patients treated with chemoradiotherapy. Methods FLT PET and MRI scans were performed before and 4 weeks after chemoradiotherapy. MRI scans were also performed after three cycles of adjuvant temozolomide. Pseudoprogression was defined as progressive disease on MRI after chemoradiotherapy with stabilisation or reduction of contrast-enhanced lesions after three cycles of temozolomide, and was compared with the disease course during long-term follow-up. Changes in maximum standardized uptake value (SUVmax) and tumour-to-normal uptake ratios were calculated for FLT and are presented as the mean SUVmax for multiple lesions. Results Between 2009 and 2012, 30 patients were included. Of 24 evaluable patients, 7 showed pseudoprogression and 7 had true progression as defined by MRI response. FLT PET parameters did not significantly differ between patients with true progression and pseudoprogression defined by MRI. The correlation between change in SUVmax and survival (p = 0.059) almost reached the standard level of statistical significance. Lower baseline FLT PET uptake was significantly correlated with improved survival (p = 0.022). Conclusion Baseline FLT uptake appears to be predictive of overall survival. Furthermore, changes in SUVmax over time showed a tendency to be associated with improved survival. However, further studies are necessary to investigate the ability of FLT PET imaging to discriminate between true progression and pseudoprogression in patients with glioblastoma.
Collapse
|
23
|
Kim YI, Kim Y, Lee JY, Jang SJ. Prognostic Value of the Metabolic and Volumetric Parameters of 11C-Methionine Positron-Emission Tomography for Gliomas: A Systematic Review and Meta-Analysis. AJNR Am J Neuroradiol 2018; 39:1629-1634. [PMID: 29954817 DOI: 10.3174/ajnr.a5707] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 03/29/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Several studies have demonstrated that 11C-methionine positron-emission tomography provides information on prognosis. PURPOSE We performed a systematic review and meta-analysis of the prognostic value of the metabolic and volumetric parameters of 11C-methionine-PET for gliomas. DATA SOURCES A systematic search was performed using the following combination of keywords: "methionine," "PET," "glioma," and "prognosis." STUDY SELECTION The inclusion criteria were the use of 11C-methionine-PET as an imaging tool, studies limited to gliomas, studies including metabolic parameters (tumor-to-normal ratio) and/or volumetric parameters (metabolic tumor volume), and studies reporting survival data. The electronic search first identified 181 records, and 14 studies were selected. DATA ANALYSIS Event-free survival and overall survival were the outcome measures of interest. The effect of the tumor-to-normal ratio and metabolic tumor volume on survival was determined by the effect size of the hazard ratio. Hazard ratios were extracted directly from each study when provided or determined by analyzing the Kaplan-Meier curves. DATA SYNTHESIS The combined hazard ratios of the tumor-to-normal ratio for event-free survival was 1.74 with no significance and that of the tumor-to-normal ratio for overall survival was 2.02 with significance. The combined hazard ratio of the metabolic tumor volume for event-free survival was 2.72 with significance and that of the metabolic tumor volume for overall survival was 3.50 with significance. LIMITATIONS The studies selected were all retrospective, and there were only 4 studies involving the metabolic tumor volume. CONCLUSIONS The present meta-analysis of 11C-methionine-PET suggests that the tumor-to-normal ratio for overall survival and the metabolic tumor volume for event-free survival and overall survival are significant prognostic factors for patients with gliomas.
Collapse
Affiliation(s)
- Y-I Kim
- From the Department of Nuclear Medicine (Y.-i.K.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Nuclear Medicine (Y.-i.K., J.Y.L., S.J.J.), CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Y Kim
- Veterans Health Service Medical Center (Y.K.), Seoul, Republic of Korea
| | - J Y Lee
- Department of Nuclear Medicine (Y.-i.K., J.Y.L., S.J.J.), CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - S J Jang
- Department of Nuclear Medicine (Y.-i.K., J.Y.L., S.J.J.), CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| |
Collapse
|
24
|
Jin T, Iordanova B, Hitchens TK, Modo M, Wang P, Mehrens H, Kim SG. Chemical exchange-sensitive spin-lock (CESL) MRI of glucose and analogs in brain tumors. Magn Reson Med 2018; 80:488-495. [PMID: 29569739 DOI: 10.1002/mrm.27183] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/02/2018] [Accepted: 02/26/2018] [Indexed: 12/27/2022]
Abstract
PURPOSE Glucose uptake and metabolism can be measured by chemical exchange-sensitive spin-lock (CESL) MRI with an administration of glucose or its analogs. This study investigates the sensitivity, the spatiotemporal characteristics, and the signal source of glucoCESL with a 9L rat brain tumor model. METHODS Dynamic CESL MRI with intravenous injection of D-glucose, 2-deoxy-D-glucose (2DG), and L-glucose were measured and compared with gadolinium-based dynamic contrast-enhanced (DCE) MRI. RESULTS The CESL signals with an injection of glucose or analogs have faster and larger changes in tumors than normal brain tissue. In tumors, the CESL signal with 2DG injection has larger and slower peak response than that with D-glucose due to the accumulation of 2DG and 2DG-6-phosphate in the intracellular compartment, whereas L-glucose, which cannot be transported intracellularly by glucose transporters, only induces a small change. The initial glucoCESL maps (< 4 minutes) are qualitatively similar to DCE maps, whereas later maps (> 4 minutes) show more widespread responses. The rise times of D-glucose-CESL and 2DG-CESL signals in the tumor are slower than that of DCE. Our data suggest that the initial CESL contrast primarily reflects a passive increase of glucose content in the extracellular space of tumors due to a higher vascular permeability, whereas the later period may have a significant contribution from the uptake/metabolism of glucose in the intracellular compartment. CONCLUSIONS Our results demonstrate that glucoCESL MRI has both extracellular and intracellular contributions, and can be a useful tool for measurements of both vascular permeability and glucose uptake in tumors.
Collapse
Affiliation(s)
- Tao Jin
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bistra Iordanova
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - T Kevin Hitchens
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michel Modo
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ping Wang
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Hunter Mehrens
- Department of Physics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Seong-Gi Kim
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea.,Department of Biomedical Engineering, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
25
|
Wang C, Li K, Li T, Chen Z, Wen Y, Liu X, Jia X, Zhang Y, Xu Y, Han M, Komatsu N, Zhao L, Chen X. Monocyte-mediated chemotherapy drug delivery in glioblastoma. Nanomedicine (Lond) 2017; 13:157-178. [PMID: 29173008 DOI: 10.2217/nnm-2017-0266] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
AIM To mechanistically prove the concept of monocyte-mediated nano drug delivery in glioblastoma (GBM). RESULTS nano-doxorubicin-loaded monocytes (Nano-DOX-MC) were viable, able to cross an artificial endothelial barrier and capable of infiltrating GBM spheroids and releasing drug therein. GBM cells stimulated unloading of Nano-DOX-MC and took up the unloaded drug and released damage-associated molecular patterns. In mice with orthotopic GBM xenografts, Nano-DOX-MC resulted in much improved tumor drug delivery efficacy and damage-associated molecular patterns emission. Mechanistically, Nano-DOX was found sequestered in the lysosomal compartment and to induce autophagy, which may underlie MC's tolerance to Nano-DOX. Lysosomal exocytosis was found involved in the discharging mechanism of intracellular Nano-DOX. CONCLUSION Nano-DOX can be effectively delivered by MC in GBM and induce cancer cell damage.
Collapse
Affiliation(s)
- Chao Wang
- Department of Pharmacology, School of Basic Medicine, Wuhan University, Donghu Avenue No.185, Wuhan 430072, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Ke Li
- Center for Lab Teaching of Basic Medicine, School of Basic Medicine, Wuhan University, Donghu Avenue No.185, Wuhan 430072, China
| | - Tongfei Li
- Department of Pharmacology, School of Basic Medicine, Wuhan University, Donghu Avenue No.185, Wuhan 430072, China
| | - Zhuo Chen
- Department of Pharmacology, School of Basic Medicine, Wuhan University, Donghu Avenue No.185, Wuhan 430072, China
| | - Yu Wen
- Department of Pharmacology, School of Basic Medicine, Wuhan University, Donghu Avenue No.185, Wuhan 430072, China
| | - Xin Liu
- Department of Pharmacology, School of Basic Medicine, Wuhan University, Donghu Avenue No.185, Wuhan 430072, China
| | - Xuemei Jia
- Department of Pharmacology, School of Basic Medicine, Wuhan University, Donghu Avenue No.185, Wuhan 430072, China
| | - Yichao Zhang
- Department of Pharmacology, School of Basic Medicine, Wuhan University, Donghu Avenue No.185, Wuhan 430072, China
| | - Yonghong Xu
- Department of Ophthalmology, Institute of Ophthalmological Research, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Min Han
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Naoki Komatsu
- Graduate School of Human & Environmental Studies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Li Zhao
- School of Radiation Medicine & Protection (SRMP), School of Radiation & Multidisciplinary Sciences (RAD-X), Medical College, Soochow University, Suzhou 215123, China
| | - Xiao Chen
- Department of Pharmacology, School of Basic Medicine, Wuhan University, Donghu Avenue No.185, Wuhan 430072, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| |
Collapse
|
26
|
Lincoln CM, Fata P, Sotardi S, Pohlen M, Uribe T, Bello JA. Imaging spectrum of immunomodulating, chemotherapeutic and radiation therapy-related intracranial effects. Br J Radiol 2017; 91:20170553. [PMID: 29039692 DOI: 10.1259/bjr.20170553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE A wide range of treatment-related side effects result in specific neurologic symptoms and signs and neuroimaging features. Even to the most seasoned neuroradiologist, elucidating therapy-related side effects from other common mimics can be challenging. We provide a pictorial survey of some common and uncommon medication-induced and therapy-related neuroimaging manifestations, discuss pathophysiology and common pitfalls in imaging and diagnosis. METHODS A case-based review is utilized to depict scenarios on a routine basis in a general radiology or neuroradiology practice such as medication-induced posterior reversible encephalopathy syndrome to the more challenging cases of pseudoprogression and pseudoregression in temozolmide and bevacizumab therapy in gliobastoma treatment protocols. CONCLUSION Knowledge of the treatment-induced imaging abnormalities is essential in the accurate interpretation and diagnosis from the most routine to most challenging of clinical situations. We provide a pictorial review for the radiologist to employ in order to be an invaluable provider to our clinical colleagues and patients.
Collapse
Affiliation(s)
- Christie M Lincoln
- 1 Department of Radiology, Baylor College of Medicine , Houston, TX , USA
| | - Peter Fata
- 2 Department of Radiology, Houston Methodist , Houston, TX , USA
| | - Susan Sotardi
- 3 Department of Neuroradiology, Massachusetts General Hospital , Boston, MA , USA
| | - Michael Pohlen
- 1 Department of Radiology, Baylor College of Medicine , Houston, TX , USA
| | - Tomas Uribe
- 1 Department of Radiology, Baylor College of Medicine , Houston, TX , USA
| | - Jacqueline A Bello
- 4 Albert Einstein College of Medicine, Montefiore Medical Center , Bronx, NY , USA
| |
Collapse
|
27
|
Villanueva-Meyer JE, Mabray MC, Cha S. Current Clinical Brain Tumor Imaging. Neurosurgery 2017; 81:397-415. [PMID: 28486641 PMCID: PMC5581219 DOI: 10.1093/neuros/nyx103] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 02/23/2017] [Indexed: 01/12/2023] Open
Abstract
Neuroimaging plays an ever evolving role in the diagnosis, treatment planning, and post-therapy assessment of brain tumors. This review provides an overview of current magnetic resonance imaging (MRI) methods routinely employed in the care of the brain tumor patient. Specifically, we focus on advanced techniques including diffusion, perfusion, spectroscopy, tractography, and functional MRI as they pertain to noninvasive characterization of brain tumors and pretreatment evaluation. The utility of both structural and physiological MRI in the post-therapeutic brain evaluation is also reviewed with special attention to the challenges presented by pseudoprogression and pseudoresponse.
Collapse
Affiliation(s)
- Javier E. Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, Neuroradiology Section, University of California San Francisco, San Francisco, California
| | - Marc C. Mabray
- Department of Radiology and Biomedical Imaging, Neuroradiology Section, University of California San Francisco, San Francisco, California
| | - Soonmee Cha
- Department of Radiology and Biomedical Imaging, Neuroradiology Section, University of California San Francisco, San Francisco, California
| |
Collapse
|
28
|
TSPO PET for glioma imaging using the novel ligand 18F-GE-180: first results in patients with glioblastoma. Eur J Nucl Med Mol Imaging 2017; 44:2230-2238. [DOI: 10.1007/s00259-017-3799-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/03/2017] [Indexed: 12/27/2022]
|
29
|
Gauvain K, Ponisio MR, Barone A, Grimaldi M, Parent E, Leeds H, Goyal M, Rubin J, McConathy J. 18F-FDOPA PET/MRI for monitoring early response to bevacizumab in children with recurrent brain tumors. Neurooncol Pract 2017; 5:28-36. [PMID: 29692922 DOI: 10.1093/nop/npx008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background Noninvasively predicting early response to therapy in recurrent pediatric brain tumors provides a challenge. 3,4-dihydroxy-6-[18F]fluoro-L-phenylalanine (18F-FDOPA) PET/MRI has not been previously studied as a tool to evaluate early response to antiangiogenic therapy in children. The purpose of this study was to evaluate the safety and feasibility of using 18F-FDOPA PET/MRI to assess response to bevacizumab in children with relapsed brain tumors. Materials and Methods Six patients with recurrent gliomas (5 low-grade, 1 high-grade) planned to undergo treatment with bevacizumab were enrolled. 18F-FDOPA PET/MRI scans were obtained prior to and 4 weeks following the start of treatment, and these were compared with the clinical response determined at the 3-month MRI. The primary PET measure was metabolic tumor volume (MTV) at 10 to 15 min after 18F-FDOPA injection. For each tumor, the MTV was determined by manually defining initial tumor volumes of interest (VOI) and then applying a 1.5-fold threshold relative to the mean standardized uptake value (SUV) of a VOI in the frontal lobe contralateral to the tumor. Results 18F-FDOPA PET/MRI was well tolerated by all patients. All tumors were well visualized with 18F-FDOPA on the initial study, with peak tumor uptake occurring approximately 10 min after injection. Maximum and mean SUVs as well as tumor-to-brain ratios were not predictors of response at 3 months. Changes in MTVs after therapy ranged from 23% to 98% (n = 5). There is a trend towards the percent MTV change seen on the 4-week scan correlating with progression-free survival. Conclusion 18F-FDOPA PET/MRI was well tolerated in pediatric patients and merits further investigation as an early predictor of response to therapy.
Collapse
Affiliation(s)
- Karen Gauvain
- Washington University School of Medicine, Pediatric Hematology/Oncology, St. Louis, MO
| | - Maria Rosana Ponisio
- Washington University School of Medicine, Pediatric Hematology/Oncology, St. Louis, MO.,Washington University School of Medicine, Mallinckrodt Institute of Radiology, St. Louis, MO
| | - Amy Barone
- Washington University School of Medicine, Pediatric Hematology/Oncology, St. Louis, MO
| | - Michael Grimaldi
- Washington University School of Medicine, Pediatric Hematology/Oncology, St. Louis, MO.,Washington University School of Medicine, Mallinckrodt Institute of Radiology, St. Louis, MO
| | - Ephraim Parent
- Washington University School of Medicine, Pediatric Hematology/Oncology, St. Louis, MO.,Washington University School of Medicine, Mallinckrodt Institute of Radiology, St. Louis, MO
| | - Hayden Leeds
- Washington University School of Medicine, Pediatric Hematology/Oncology, St. Louis, MO.,Washington University School of Medicine, Mallinckrodt Institute of Radiology, St. Louis, MO
| | - Manu Goyal
- Washington University School of Medicine, Pediatric Hematology/Oncology, St. Louis, MO.,Washington University School of Medicine, Mallinckrodt Institute of Radiology, St. Louis, MO
| | - Joshua Rubin
- Washington University School of Medicine, Pediatric Hematology/Oncology, St. Louis, MO
| | - Jonathan McConathy
- Washington University School of Medicine, Pediatric Hematology/Oncology, St. Louis, MO.,University of Alabama at Birmingham, Department of Radiology, Birmingham, AL
| |
Collapse
|
30
|
Filss CP, Cicone F, Shah NJ, Galldiks N, Langen KJ. Amino acid PET and MR perfusion imaging in brain tumours. Clin Transl Imaging 2017; 5:209-223. [PMID: 28680873 PMCID: PMC5487907 DOI: 10.1007/s40336-017-0225-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 02/28/2017] [Indexed: 12/17/2022]
Abstract
Purpose Despite the excellent capacity of the conventional MRI to image brain tumours, problems remain in answering a number of critical diagnostic questions. To overcome these diagnostic shortcomings, PET using radiolabeled amino acids and perfusion-weighted imaging (PWI) are currently under clinical evaluation. The role of amino acid PET and PWI in different diagnostic challenges in brain tumours is controversial. Methods Based on the literature and experience of our centres in correlative imaging with PWI and PET using O-(2-[18F]fluoroethyl)-l-tyrosine or 3,4-dihydroxy-6-[18F]-fluoro-l-phenylalanine, the current role and shortcomings of amino acid PET and PWI in different diagnostic challenges in brain tumours are reviewed. Literature searches were performed on PubMed, and additional literature was retrieved from the reference lists of identified articles. In particular, all studies in which amino acid PET was directly compared with PWI were included. Results PWI is more readily available, but requires substantial expertise and is more sensitive to artifacts than amino acid PET. At initial diagnosis, PWI and amino acid PET can help to define a site for biopsy but amino acid PET appears to be more powerful to define the tumor extent. Both methods are helpful to differentiate progression or recurrence from unspecific posttherapeutic changes. Assessment of therapeutic efficacy can be achieved especially with amino acid PET, while the data with PWI are sparse. Conclusion Both PWI and amino acid PET add valuable diagnostic information to the conventional MRI in the assessment of patients with brain tumours, but further studies are necessary to explore the complementary nature of these two methods.
Collapse
Affiliation(s)
- Christian P Filss
- Institute of Neuroscience and Medicine (INM-3, INM-4), Forschungszentrum Jülich, Jülich, Germany.,Departments of Nuclear Medicine and Neurology, RWTH Aachen University Clinic, Aachen, Germany
| | - Francesco Cicone
- Unit of Nuclear Medicine, Department of Surgical and Medical Sciences and Translational Medicine, Sapienza University of Rome, Rome, Italy.,Nuclear Medicine and Molecular Medicine Department, University Hospital of Lausanne, Lausanne, Switzerland
| | - Nadim Jon Shah
- Institute of Neuroscience and Medicine (INM-3, INM-4), Forschungszentrum Jülich, Jülich, Germany.,Departments of Nuclear Medicine and Neurology, RWTH Aachen University Clinic, Aachen, Germany.,JARA-Jülich Aachen Research Alliance, Jülich, Germany.,Monash Institute of Medical Engineering, Department of Electrical and Computer Systems Engineering, and Monash Biomedical Imaging, School of Psychological Sciences, Monash University, Melbourne, VIC Australia
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3, INM-4), Forschungszentrum Jülich, Jülich, Germany.,Department of Neurology, University of Cologne, Cologne, Germany.,Center of Integrated Oncology (CIO), University of Cologne and Bonn, Cologne, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, INM-4), Forschungszentrum Jülich, Jülich, Germany.,Departments of Nuclear Medicine and Neurology, RWTH Aachen University Clinic, Aachen, Germany.,JARA-Jülich Aachen Research Alliance, Jülich, Germany
| |
Collapse
|
31
|
Stegmayr C, Oliveira D, Niemietz N, Willuweit A, Lohmann P, Galldiks N, Shah NJ, Ermert J, Langen KJ. Influence of Bevacizumab on Blood-Brain Barrier Permeability and O-(2- 18F-Fluoroethyl)-l-Tyrosine Uptake in Rat Gliomas. J Nucl Med 2017; 58:700-705. [PMID: 28153956 DOI: 10.2967/jnumed.116.187047] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 12/26/2016] [Indexed: 01/20/2023] Open
Abstract
Restoration of the blood-brain barrier (BBB) after antiangiogenic therapy of gliomas with bevacizumab may result in a decrease in contrast enhancement on MRI despite tumor progression. This so-called pseudoresponse is difficult to differentiate from a true tumor response with conventional MRI. Initial patient studies have indicated that PET using O-(2-18F-fluoroethyl)-l-tyrosine (18F-FET) may be helpful for solving this diagnostic problem. This study was performed to investigate the effects of bevacizumab on BBB permeability and 18F-FET uptake in a human xenograft model. Methods: Human U87 glioblastoma cells were implanted into the striatum of immunodeficient RNU rats. 18F-FET PET scans and ex vivo autoradiography were performed in animals receiving a single high dose of bevacizumab (45 mg/kg 2 d before PET; n = 9) or in animals receiving 2 lower doses (10 mg/kg 9 and 2 d before PET; n = 10) to evaluate short-term and long-term effects on the BBB, respectively, and in control animals without bevacizumab treatment (n = 8). Time-activity curves, slope, and tumor-to-brain ratios of 18F-FET uptake (18-61 min after injection) were evaluated using a volume-of-interest analysis. After PET scanning, Evans blue dye (EBD) was injected into animals, and cryosections of the brains were evaluated by autoradiography, by histology, and for EBD fluorescence to assess BBB permeability. Results: Compared with the control, short-term bevacizumab therapy resulted in a trend toward BBB restoration (P = 0.055) and long-term therapy resulted in a significant decrease (P = 0.004) in BBB permeability, as assessed by EBD fluorescence. In contrast, no significant differences in tumor-to-brain ratios or slope of 18F-FET uptake were observed in PET and autoradiography (P > 0.05). Conclusion:8F-FET uptake in glioblastomas seems to be largely independent of BBB permeability and reflects the viability of tumor tissue during antiangiogenic therapy more reliably than contrast-enhanced MRI.
Collapse
Affiliation(s)
- Carina Stegmayr
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
| | - Dennis Oliveira
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
| | - Nicole Niemietz
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
| | - Antje Willuweit
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany.,Department of Neurology, University of Cologne, Cologne, Germany
| | - N Jon Shah
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany.,Department of Neurology, RWTH University, Aachen, Germany.,Jülich-Aachen Research Alliance (JARA)-Section JARA-Brain, RWTH Aachen University, Aachen, Germany; and
| | - Johannes Ermert
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany.,Department of Nuclear Medicine, RWTH University Hospital, Aachen, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5, INM-11), Forschungszentrum Jülich, Jülich, Germany .,Department of Nuclear Medicine, RWTH University Hospital, Aachen, Germany
| |
Collapse
|
32
|
Yamamoto J, Kakeda S, Yoneda T, Ogura SI, Shimajiri S, Tanaka T, Korogi Y, Nishizawa S. Improving contrast enhancement in magnetic resonance imaging using 5-aminolevulinic acid-induced protoporphyrin IX for high-grade gliomas. Oncol Lett 2016; 13:1269-1275. [PMID: 28454245 DOI: 10.3892/ol.2016.5539] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 10/28/2016] [Indexed: 12/13/2022] Open
Abstract
Magnetic resonance imaging (MRI) with a gadolinium-based contrast agent is the gold standard for high-grade gliomas (HGGs). The compound 5-aminolevulinic acid (5-ALA) undergoes a high rate of cellular uptake, particularly in cancer cells. In addition, fluorescence-guided resection with 5-ALA is widely used for imaging HGGs. 5-ALA is water soluble, while protoporphyrin IX (PpIX) is water insoluble. It was speculated whether converting from 5-ALA to PpIX may relatively increase intracellular water content, and consequently, might enhance the T2 signal intensity in HGG. The aim of the present study was to assess whether 5-ALA-induced PpIX enhances the T2 signal intensity in patients with HGGs. A total of 4 patients who were candidates for HGG surgical treatment were prospectively analyzed with preoperative MRI. Patients received oral doses of 5-ALA (20 mg/kg) 3 h prior to anesthesia. At 2.5 h post-5-ALA administration, T2-weighted images (T2WIs) were obtained from all patients. Subsequently, tumors were evaluated via fluorescence using a modified operating microscope. Fluorescent tumor tissues were obtained to analyze the accumulation of 5-ALA-induced PpIX within the tumors, which was confirmed quantitatively by high-performance liquid chromatography (HPLC) analysis. The MRI T2 signal intensity within the tumors was evaluated prior to and following 5-ALA administration. Three glioblastoma multiformes (GBMs) and 1 anaplastic oligodendroglioma (AO) were included in the analysis. Intraoperatively, all GBMs exhibited strong fluorescence of 5-ALA-induced PpIX, whilst no fluorescence was observed in the AO sample. HPLC analysis indicated a higher accumulation of 5-ALA-induced PpIX in the GBM samples compared with the AO sample. In total, 48 regions of interest were identified within the tumors from T2-WIs. In the GBM group, the relative T2 signal intensity value within the tumors following 5-ALA administration was significantly increased compared with the T2 signal intensity value prior to 5-ALA administration (1.537±0.021 and 1.577±0.023, respectively; P=0.0055). No significant differences were observed in the AO group. These results suggest that the 5-ALA-induced PpIX enhanced the T2 signal intensity in HGG. Therefore, 5-ALA may be a potentially useful MRI contrast reagent for HGG.
Collapse
Affiliation(s)
- Junkoh Yamamoto
- Department of Neurosurgery, University of Occupational and Environmental Health, Kitakyushu, Fukuoka 807-8555, Japan
| | - Shingo Kakeda
- Department of Radiology, University of Occupational and Environmental Health, Kitakyushu, Fukuoka 807-8555, Japan
| | - Tetsuya Yoneda
- Department of Medical Physics in Advanced Biomedical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0976, Japan
| | - Shun-Ichiro Ogura
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Shohei Shimajiri
- Department of Surgical Pathology, University of Occupational and Environmental Health, Kitakyushu, Fukuoka 807-8555, Japan
| | | | - Yukunori Korogi
- Department of Radiology, University of Occupational and Environmental Health, Kitakyushu, Fukuoka 807-8555, Japan
| | - Shigeru Nishizawa
- Department of Neurosurgery, University of Occupational and Environmental Health, Kitakyushu, Fukuoka 807-8555, Japan
| |
Collapse
|
33
|
Change in 18F-Fluoromisonidazole PET Is an Early Predictor of the Prognosis in the Patients with Recurrent High-Grade Glioma Receiving Bevacizumab Treatment. PLoS One 2016; 11:e0167917. [PMID: 27936194 PMCID: PMC5148016 DOI: 10.1371/journal.pone.0167917] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 11/22/2016] [Indexed: 11/19/2022] Open
Abstract
Background Bevacizumab (BEV), a humanized monoclonal antibody, become a currently important chemotherapeutic option for the patients with recurrent glioma. The aim of this retrospective study is to investigate whether 18F-Fluoromisonidazole (FMISO) PET have the potential to detect BEV-resistant gliomas in the early-stage. Methods We reviewed the FMISO PET and MRI appearances before and 3 to 4 courses after BEV treatment on 18 recurrent glioma patients. FMISO accumulation was assessed by visual inspection and semi-quantitative values which were tumor-to-normal (T/N) ratio and hypoxic volume. MRI responses were evaluated based on RANO (Response Assessment in Neuro-Oncology) criteria. The prognostic analysis was performed in relation to the response assessment by FMISO PET and MRI using overall survival (OS) after BEV application. Results After BEV application, MRI revealed partial response in 14 of 18 patients (78%), of which 9 patients also demonstrated decreased FMISO accumulation. These 9 patients (50%) were classified as “MRI-FMISO double responder”. As for the other 5 patients (28%), FMISO accumulation volumes increased or remained stable after BEV treatment although partial responses were achieved on MRI. Therefore, these cases were classified as “MRI-only responder”. The remaining 4 patients (22%) did not show treatment response on FMISO PET or MRI (“non-responder”). MRI-FMISO double responders showed significantly longer OS than that in other groups (median 12.4 vs 5.7 months; P < 0.001), whereas there were no overall survival difference between MRI-only responders and non-responders (median OS, 5.7 and 4.8 months; P = 0.58). Among the pre-treatment clinical factors, high FMISO T/N ratio was a significant prognostic factor of overall survival in these patients under the assessment of Cox proportional hazard model. Conclusions Recurrent gliomas with decreasing FMISO accumulation after short-term BEV application could derive a survival benefit from BEV treatment. Change in FMISO PET appearance can identify BEV-resistant gliomas in early-stage regardless of MRI findings in a comprehensible way.
Collapse
|
34
|
Lyu Y, Liu S, You H, Hou B, Wang Y, Ma W, Feng F. Evaluation of recurrent high-grade gliomas treated with bevacizumab: A preliminary report of 3D pseudocontinuous artery spin labeling. J Magn Reson Imaging 2016; 46:565-573. [PMID: 27902863 DOI: 10.1002/jmri.25558] [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: 04/12/2016] [Accepted: 11/01/2016] [Indexed: 12/14/2022] Open
Abstract
PURPOSE To investigate the role of cerebral blood flow (CBF) derived from a 3D fast spin echo (FSE) pseudocontinuous artery spin labeling (pcASL) sequence in evaluating the survival rate of recurrent high-grade gliomas (rHGGs) that were treated with bevacizumab (BEV). MATERIALS AND METHODS Sixteen patients with rHGGs who underwent 3T 3D FSE pcASL imaging 1-2 days before (baseline or pre-BEV) and within 1 month after BEV treatment initiation (post-BEV) were included in the study. Average (aCBF) and maximum (mCBF) cerebral blood flow of the enhancing tumor, their respective normalized values to contralateral normal-appearing white matter (rCBF_wm and mCBF_wm) and cerebellum (rCBF_cb and mCBF_cb), and the related changes between baseline and post-BEV were evaluated. Receiver operating characteristic (ROC) curve analysis was utilized to define the optimal cutoff perfusion values for overall survival (OS) and progression-free survival (PFS) stratification. Kaplan-Meier analysis with log-rank test was applied to assess and compare PFS and OS rates. RESULTS All the CBF measurements pre-BEV and post-BEV treatment were significantly different except mCBF. The CBF measurements (aCBF, rCBF_wm, rCBF_cb, mCBF_wm and mCBF_cb) pre-BEV all decreased post-BEV treatment. Cutoffs of aCBF (43.72 ml/100g/min) pre-BEV for OS, rCBF_cb (1.09) pre-BEV for PFS and OS, and ΔaCBF (-0.37) for PFS were found to be statistically significant in survival stratification (404 days vs. 140 days, P = 0.026; 251 days vs. 112 days, P = 0.044; 404 days vs. 194 days, P = 0.046; 267 days vs. 116 days, P = 0.048, respectively). CONCLUSION Three dimensional FSE pcASL can detect the decrease of perfusion in rHGGs treated with BEV and is a potential promising technique in stratifying survival rate of rHGGs under BEV treatment. LEVEL OF EVIDENCE 3 Technical Efficacy: Stage 2 J. MAGN. RESON. IMAGING 2017;46:565-573.
Collapse
Affiliation(s)
- Yuelei Lyu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Wangfujing Dongcheng District, Beijing, P.R. China
| | - Shuai Liu
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Wangfujing Dongcheng District, Beijing, P.R. China
| | - Hui You
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Wangfujing Dongcheng District, Beijing, P.R. China
| | - Bo Hou
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Wangfujing Dongcheng District, Beijing, P.R. China
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Wangfujing Dongcheng District, Beijing, P.R. China
| | - Wenbin Ma
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Wangfujing Dongcheng District, Beijing, P.R. China
| | - Feng Feng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Wangfujing Dongcheng District, Beijing, P.R. China
| |
Collapse
|
35
|
MRI and 11C-methyl-L-methionine PET Differentiate Bevacizumab True Responders After Initiating Therapy for Recurrent Glioblastoma. Clin Nucl Med 2016; 41:852-857. [DOI: 10.1097/rlu.0000000000001377] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
36
|
Amino-acid PET versus MRI guided re-irradiation in patients with recurrent glioblastoma multiforme (GLIAA) - protocol of a randomized phase II trial (NOA 10/ARO 2013-1). BMC Cancer 2016; 16:769. [PMID: 27716184 PMCID: PMC5052714 DOI: 10.1186/s12885-016-2806-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 09/22/2016] [Indexed: 12/21/2022] Open
Abstract
Background The higher specificity of amino-acid positron emission tomography (AA-PET) in the diagnosis of gliomas, as well as in the differentiation between recurrence and treatment-related alterations, in comparison to contrast enhancement in T1-weighted MRI was demonstrated in many studies and is the rationale for their implementation into radiation oncology treatment planning. Several clinical trials have demonstrated the significant differences between AA-PET and standard MRI concerning the definition of the gross tumor volume (GTV). A small single-center non-randomized prospective study in patients with recurrent high grade gliomas treated with stereotactic fractionated radiotherapy (SFRT) showed a significant improvement in survival when AA-PET was integrated in target volume delineation, in comparison to patients treated based on CT/MRI alone. Methods This protocol describes a prospective, open label, randomized, multi-center phase II trial designed to test if radiotherapy target volume delineation based on FET-PET leads to improvement in progression free survival (PFS) in patients with recurrent glioblastoma (GBM) treated with re-irradiation, compared to target volume delineation based on T1Gd-MRI. The target sample size is 200 randomized patients with a 1:1 allocation ratio to both arms. The primary endpoint (PFS) is determined by serial MRI scans, supplemented by AA-PET-scans and/or biopsy/surgery if suspicious of progression. Secondary endpoints include overall survival (OS), locally controlled survival (time to local progression or death), volumetric assessment of GTV delineated by either method, topography of progression in relation to MRI- or PET-derived target volumes, rate of long term survivors (>1 year), localization of necrosis after re-irradiation, quality of life (QoL) assessed by the EORTC QLQ-C15 PAL questionnaire, evaluation of safety of FET-application in AA-PET imaging and toxicity of re-irradiation. Discussion This is a protocol of a randomized phase II trial designed to test a new strategy of radiotherapy target volume delineation for improving the outcome of patients with recurrent GBM. Moreover, the trial will help to develop a standardized methodology for the integration of AA-PET and other imaging biomarkers in radiation treatment planning. Trial registration The GLIAA trial is registered with ClinicalTrials.gov (NCT01252459, registration date 02.12.2010), German Clinical Trials Registry (DRKS00000634, registration date 10.10.2014), and European Clinical Trials Database (EudraCT-No. 2012-001121-27, registration date 27.02.2012).
Collapse
|
37
|
Molina D, Pérez-Beteta J, Martínez-González A, Sepúlveda JM, Peralta S, Gil-Gil MJ, Reynes G, Herrero A, De Las Peñas R, Luque R, Capellades J, Balaña C, Pérez-García VM. Geometrical Measures Obtained from Pretreatment Postcontrast T1 Weighted MRIs Predict Survival Benefits from Bevacizumab in Glioblastoma Patients. PLoS One 2016; 11:e0161484. [PMID: 27557121 PMCID: PMC4996463 DOI: 10.1371/journal.pone.0161484] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/06/2016] [Indexed: 11/18/2022] Open
Abstract
Background Antiangiogenic therapies for glioblastoma (GBM) such as bevacizumab (BVZ), have been unable to extend survival in large patient cohorts. However, a subset of patients having angiogenesis-dependent tumors might benefit from these therapies. Currently, there are no biomarkers allowing to discriminate responders from non-responders before the start of the therapy. Methods 40 patients from the randomized GENOM009 study complied the inclusion criteria (quality of images, clinical data available). Of those, 23 patients received first line temozolomide (TMZ) for eight weeks and then concomitant radiotherapy and TMZ. 17 patients received BVZ+TMZ for seven weeks and then added radiotherapy to the treatment. Clinical variables were collected, tumors segmented and several geometrical measures computed including: Contrast enhancing (CE), necrotic, and total volumes; equivalent spherical CE width; several geometric measures of the CE ‘rim’ geometry and a set of image texture measures. The significance of the results was studied using Kaplan-Meier and Cox proportional hazards analysis. Correlations were assessed using Spearman correlation coefficients. Results Kaplan-Meier and Cox proportional hazards analysis showed that total, CE and inner volume (p = 0.019, HR = 4.258) and geometric heterogeneity of the CE areas (p = 0.011, HR = 3.931) were significant parameters identifying response to BVZ. The group of patients with either regular CE areas (small geometric heterogeneity, median difference survival 15.88 months, p = 0.011) or those with small necrotic volume (median survival difference 14.50 months, p = 0.047) benefited substantially from BVZ. Conclusion Imaging biomarkers related to the irregularity of contrast enhancing areas and the necrotic volume were able to discriminate GBM patients with a substantial survival benefit from BVZ. A prospective study is needed to validate our results.
Collapse
Affiliation(s)
- David Molina
- Laboratory of Mathematical Oncology (MôLAB), Instituto de Matemática Aplicada a la Ciencia y la Ingeniería, Edificio Politécnico, Avda. Camilo José Cela 3, Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain
- * E-mail:
| | - Julián Pérez-Beteta
- Laboratory of Mathematical Oncology (MôLAB), Instituto de Matemática Aplicada a la Ciencia y la Ingeniería, Edificio Politécnico, Avda. Camilo José Cela 3, Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - Alicia Martínez-González
- Laboratory of Mathematical Oncology (MôLAB), Instituto de Matemática Aplicada a la Ciencia y la Ingeniería, Edificio Politécnico, Avda. Camilo José Cela 3, Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - Juan M. Sepúlveda
- Medical Oncology Service, Hospital Universitario, 12 de Octubre, Madrid, Spain
| | - Sergi Peralta
- Medical Oncology Service, Hospital Sant Joan de Reus, Reus, Spain
| | - Miguel J. Gil-Gil
- Medical Oncology Service, Institut Catalá d’Oncologia IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
| | - Gaspar Reynes
- Medical Oncology Service, Hospital Universitario La Fe, Valencia, Spain
| | - Ana Herrero
- Medical Oncology Service, Hospital Miguel Servet, Zaragoza, Spain
| | - Ramón De Las Peñas
- Medical Oncology Service, Hospital Provincial de Castellón, Castellón, Spain
| | - Raquel Luque
- Medical Oncology Service, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Jaume Capellades
- Neuroradiology Section. Radiology Service. Hospital del Mar, Barcelona, Spain
| | - Carmen Balaña
- Medical Oncology Service, Institut Català d’Oncologia, IGTP, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Víctor M. Pérez-García
- Laboratory of Mathematical Oncology (MôLAB), Instituto de Matemática Aplicada a la Ciencia y la Ingeniería, Edificio Politécnico, Avda. Camilo José Cela 3, Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain
| |
Collapse
|
38
|
Oehlke O, Grosu AL. PET/MRI and brain tumors: focus on radiation oncology treatment planning. Clin Transl Imaging 2016. [DOI: 10.1007/s40336-016-0206-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
39
|
Miyake K, Ogawa D, Okada M, Hatakeyama T, Tamiya T. Usefulness of positron emission tomographic studies for gliomas. Neurol Med Chir (Tokyo) 2016; 56:396-408. [PMID: 27250577 PMCID: PMC4945598 DOI: 10.2176/nmc.ra.2015-0305] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Non-invasive positron emission tomography (PET) enables the measurement of metabolic and molecular processes with high sensitivity. PET plays a significant role in the diagnosis, prognosis, and treatment of brain tumors and predominantly detects brain tumors by detecting their metabolic alterations, including energy metabolism, amino acids, nucleic acids, and hypoxia. Glucose metabolic tracers are related to tumor cell energy and exhibit good sensitivity but poor specificity for malignant tumors. Amino acid metabolic tracers provide a better delineation of tumors and cellular proliferation. Nucleic acid metabolic tracers have a high sensitivity for malignant tumors and cellular proliferation. Hypoxic metabolism tracers are useful for detecting resistance to radiotherapy and chemotherapy. Therefore, PET imaging techniques are useful for detecting biopsy-targeting points, deciding on tumor resection, radiotherapy planning, monitoring therapy, and distinguishing brain tumor recurrence or progression from post-radiotherapy effects. However, it is not possible to use only one PET tracer to make all clinical decisions because each tracer has both advantages and disadvantages. This study focuses on the different kinds of PET tracers and summarizes their recent applications in patients with gliomas. Combinational uses of PET tracers are expected to contribute to differential diagnosis, prognosis, treatment targeting, and monitoring therapy.
Collapse
Affiliation(s)
- Keisuke Miyake
- Department of Neurological Surgery, Kagawa University Faculty of Medicine
| | | | | | | | | |
Collapse
|
40
|
Yue X, Cao D, Lan F, Pan Q, Xia T, Yu H. MiR-301a is activated by the Wnt/β-catenin pathway and promotes glioma cell invasion by suppressing SEPT7. Neuro Oncol 2016; 18:1288-96. [PMID: 27006177 DOI: 10.1093/neuonc/now044] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 02/19/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND miR-301a is frequently dysregulated and specific to human tumors, playing a critical role in tumorigenesis; however, the exact functions and regulatory mechanisms of miR-301a in glioma cells remain largely unknown. Herein, we show that miR-301a activated by the Wnt/β-catenin pathway promoted the invasion of glioma cells by directly targeting SEPT7. METHODS Biochemical, luciferase reporter, and hromatin immunoprecipitation PCR assays characterized the function and regulatory mechanisms of miR-301a in glioma invasion. RESULTS Initially, we detected the expression of miR-301a in glioma tissues and identified that miR-301a had increased, with ascending grades of the tumor. Furthermore, high levels of miR-301a were associated with a poorer prognosis in glioma patients. It is important to note that the Wnt/β-catenin/TCF4 pathway enhanced miR-301a expression by binding to the promoter region. To determine the oncogenic functions of miR-301a in glioma, SEPT7 was supported as the direct target gene. In addition, the Wnt/β-catenin pathway repressed SEPT7 expression, which was dependent on miR-301a in glioma cells. Finally, miR-301a was activated by Wnt/β-catenin and then promoted invasion of glioma cells by inhibiting the expression of SEPT7 in vitro and in vivo. CONCLUSIONS Our findings revealed the mechanism of action for miR-301a in tumor cell invasion. Moreover, the Wnt/miR-301a/SEPT7 signaling axis might be a novel target in treating glioma.
Collapse
Affiliation(s)
- Xiao Yue
- Tianjin Huanhu Hospital, Tianjin Neurosurgery Institute, Tianjin , China (X.Y., D.C.); Department of Radiation Oncology, Tianjin Hospital, Tianjin , China (F.L.); Department of Neurosurgery, Laiwu City People's Hospital, Laiwu, China (Q.P.); Department of Radiation Oncology, P.L.A Airforce General Hospital, Beijing, China (T.X.); Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiotherapy, Peking University Cancer Hospital & Institute, Beijing, China (H.Y.)
| | - Dechen Cao
- Tianjin Huanhu Hospital, Tianjin Neurosurgery Institute, Tianjin , China (X.Y., D.C.); Department of Radiation Oncology, Tianjin Hospital, Tianjin , China (F.L.); Department of Neurosurgery, Laiwu City People's Hospital, Laiwu, China (Q.P.); Department of Radiation Oncology, P.L.A Airforce General Hospital, Beijing, China (T.X.); Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiotherapy, Peking University Cancer Hospital & Institute, Beijing, China (H.Y.)
| | - FengMing Lan
- Tianjin Huanhu Hospital, Tianjin Neurosurgery Institute, Tianjin , China (X.Y., D.C.); Department of Radiation Oncology, Tianjin Hospital, Tianjin , China (F.L.); Department of Neurosurgery, Laiwu City People's Hospital, Laiwu, China (Q.P.); Department of Radiation Oncology, P.L.A Airforce General Hospital, Beijing, China (T.X.); Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiotherapy, Peking University Cancer Hospital & Institute, Beijing, China (H.Y.)
| | - Qiang Pan
- Tianjin Huanhu Hospital, Tianjin Neurosurgery Institute, Tianjin , China (X.Y., D.C.); Department of Radiation Oncology, Tianjin Hospital, Tianjin , China (F.L.); Department of Neurosurgery, Laiwu City People's Hospital, Laiwu, China (Q.P.); Department of Radiation Oncology, P.L.A Airforce General Hospital, Beijing, China (T.X.); Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiotherapy, Peking University Cancer Hospital & Institute, Beijing, China (H.Y.)
| | - Tingyi Xia
- Tianjin Huanhu Hospital, Tianjin Neurosurgery Institute, Tianjin , China (X.Y., D.C.); Department of Radiation Oncology, Tianjin Hospital, Tianjin , China (F.L.); Department of Neurosurgery, Laiwu City People's Hospital, Laiwu, China (Q.P.); Department of Radiation Oncology, P.L.A Airforce General Hospital, Beijing, China (T.X.); Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiotherapy, Peking University Cancer Hospital & Institute, Beijing, China (H.Y.)
| | - Huiming Yu
- Tianjin Huanhu Hospital, Tianjin Neurosurgery Institute, Tianjin , China (X.Y., D.C.); Department of Radiation Oncology, Tianjin Hospital, Tianjin , China (F.L.); Department of Neurosurgery, Laiwu City People's Hospital, Laiwu, China (Q.P.); Department of Radiation Oncology, P.L.A Airforce General Hospital, Beijing, China (T.X.); Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiotherapy, Peking University Cancer Hospital & Institute, Beijing, China (H.Y.)
| |
Collapse
|
41
|
Xiao Y, Zhang L, Song Z, Guo C, Zhu J, Li Z, Zhu S. Potential Diagnostic and Prognostic Value of Plasma Circulating MicroRNA-182 in Human Glioma. Med Sci Monit 2016; 22:855-62. [PMID: 26978735 PMCID: PMC4795091 DOI: 10.12659/msm.897164] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Background Previous studies showed the aberrant expression of microRNA-182 (miR-182) in glioma tissue. However, the exact role of circulating miR-182 in glioma remains unclear. Here, we confirmed the expression of plasma circulating miR-182 in glioma patients, and further explored its potential diagnostic and prognostic value. Material/Methods Real-time quantitative PCR (RT-PCR) was used to measure circulating cell-free miR-182 from 112 glioma patients and 54 healthy controls. Results Our findings showed that the level of circulating miR-182 in glioma patients was higher than that in healthy controls (P<0.001), which was significantly associated with KPS score (P=0.025) and WHO grade (P<0.001). The area under the receiver operating characteristic (ROC) curve (AUC) was 0.778. The optimal cut-off value was 1.56, and the sensitivity and specificity were 58.5% and 85.2%, respectively. Interestingly, a high predictive value of circulating miR-182 was observed in high-grade glioma (AUC=0.815). However, the AUC was lower in low-grade glioma (AUC=0.621). Kaplan-Meier analysis demonstrated that the cumulative 5-year overall survival rate in the high miR-182 group was significantly lower than that in the low miR-182 group in both overall survival (OS) (P=0.003) and disease-free survival (DFS) (P=0.006). Moreover, multivariate Cox analysis revealed that circulating miR-182 was an independent prognostic indicator for OS (P=0.034) and DFS (P=0.013). Conclusions These results suggest that circulating miR-182 may be a potential noninvasive biomarker for the diagnosis and prognosis of human glioma.
Collapse
Affiliation(s)
- Yilei Xiao
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Lina Zhang
- Department of Intensive Care Medicine, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| | - Zikun Song
- Department of Intensive Care Medicine, The People's Second Hospital of Liaocheng, Linqing, Shandong, China (mainland)
| | - Chuanjun Guo
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| | - Jianxin Zhu
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| | - Zhongmin Li
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| | - Shugan Zhu
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
42
|
Kinoshita M, Arita H, Okita Y, Kagawa N, Kishima H, Hashimoto N, Tanaka H, Watanabe Y, Shimosegawa E, Hatazawa J, Fujimoto Y, Yoshimine T. Comparison of diffusion tensor imaging and 11C-methionine positron emission tomography for reliable prediction of tumor cell density in gliomas. J Neurosurg 2016; 125:1136-1142. [PMID: 26918477 DOI: 10.3171/2015.11.jns151848] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Diffusion MRI is attracting increasing interest for tissue characterization of gliomas, especially after the introduction of antiangiogenic therapy to treat malignant gliomas. The goal of the current study is to elucidate the actual magnitude of the correlation between diffusion MRI and cell density within the tissue. The obtained results were further extended and compared with metabolic imaging with 11C-methionine (MET) PET. METHODS Ninety-eight tissue samples from 37 patients were stereotactically obtained via an intraoperative neuronavigation system. Diffusion tensor imaging (DTI) and MET PET were performed as routine presurgical imaging studies for these patients. DTI was converted into fractional anisotropy (FA) and apparent diffusion coefficient (ADC) maps, and MET PET images were registered to Gd-administered T1-weighted images that were used for navigation. Metrics of FA, ADC, and tumor-to-normal tissue ratio of MET PET along with relative values of FA (rFA) and ADC (rADC) compared with normal-appearing white matter were correlated with cell density of the stereotactically obtained tissues. RESULTS rADC was significantly lower in lesions obtained from Gd-enhancing lesions than from nonenhancing lesions. Although rADC showed a moderate but statistically significant negative correlation with cell density (p = 0.010), MET PET showed a superb positive correlation with cell density (p < 0.0001). On the other hand, rFA showed little correlation with cell density. CONCLUSIONS The presented data validated the use of rADC for estimating the treatment response of gliomas but also caution against overestimating its limited accuracy compared with MET PET.
Collapse
Affiliation(s)
- Manabu Kinoshita
- Department of Neurosurgery, Osaka Medical Center for Cancer and Cardiovascular Diseases;,Departments of 2 Neurosurgery
| | | | - Yoshiko Okita
- Department of Neurosurgery, Osaka National Hospital, National Hospital Organization, Osaka, Japan
| | | | | | | | | | | | - Eku Shimosegawa
- Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine; and
| | - Jun Hatazawa
- Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine; and
| | | | | |
Collapse
|
43
|
Positron emission tomography of high-grade gliomas. J Neurooncol 2016; 127:415-25. [PMID: 26897013 DOI: 10.1007/s11060-016-2077-1] [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: 11/06/2015] [Accepted: 02/15/2016] [Indexed: 10/22/2022]
Abstract
High-grade gliomas [HGG (WHO grades III-IV)] are almost invariably fatal. Imaging of HGG is important for orientating diagnosis, prognosis and treatment planning and is crucial for development of novel, more effective therapies. Given the potentially unlimited number of usable tracing molecules and the elevated number of available radionuclides, PET allows gathering multiple informations on HGG including data on tissue metabolism and drug pharmacokinetics. PET studies on the diagnosis, prognosis and treatment of HGG carried out by most frequently used tracers and radionuclides ((11)C and (18)F) and published in 2014 have been reviewed. These studies demonstrate that a thorough choice of tracers may confer elevated diagnostic and prognostic power to PET imaging of HGG. They also suggest that a combination of PET and MRI may give the most complete and reliable imaging information on HGG and that research on hybrid PET/MRI may be paying back in terms of improved diagnosis, prognosis and treatment planning of these deadly tumours.
Collapse
|
44
|
Suchorska B, Albert NL, Tonn JC. Usefulness of PET Imaging to Guide Treatment Options in Gliomas. Curr Treat Options Neurol 2016; 18:4. [PMID: 26815310 DOI: 10.1007/s11940-015-0384-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OPINION STATEMENT Magnetic resonance imaging (MRI) is the gold standard guiding diagnostic and therapeutic management in glioma with its high resolution and possibility to depict blood-brain-barrier disruption when contrast medium is applied. In light of the shifting paradigms revealing distinct tumor subtypes based on the molecular and genetic characterization and increasing knowledge about the variability of glioma biology, additional imaging modalities such as positron emission tomography (PET) depicting metabolic processes gain further importance in the management of glioma.
Collapse
Affiliation(s)
- Bogdana Suchorska
- Department of Neurosurgery, University Hospital Munich, Marchioninistr. 15, 81377, Munich, Germany.
| | | | - Jörg-Christian Tonn
- Department of Neurosurgery, University Hospital Munich, Marchioninistr. 15, 81377, Munich, Germany
| |
Collapse
|