1
|
Krause S, Florea A, Choi CH, Worthoff WA, Heinzel A, Fischer S, Burda N, Neumaier B, Shah NJ, Lohmann P, Mottaghy FM, Langen KJ, Stegmayr C. Autoradiography of Intracerebral Tumours in the Chick Embryo Model: A Feasibility Study Using Different PET Tracers. Mol Imaging Biol 2025; 27:151-162. [PMID: 39838234 PMCID: PMC12062108 DOI: 10.1007/s11307-025-01983-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/19/2024] [Accepted: 01/03/2025] [Indexed: 01/23/2025]
Abstract
PURPOSE In addition to rodent models, the chick embryo model has gained attention for radiotracer evaluation. Previous studies have investigated tumours on the chorioallantoic membrane (CAM), but its value for radiotracer imaging of intracerebral tumours has yet to be demonstrated. PROCEDURES Human U87 glioblastoma cells and U87-IDH1 mutant glioma cells were implanted into the brains of chick embryos at developmental day 5. After 12-14 days of tumour growth, blood-brain-barrier integrity was evaluated in vivo using MRI contrast enhancement or ex vivo with Evans blue dye. The tracers O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) (n = 5), 3,4-dihydroxy-6-[18F]-fluoro-L-phenylalanine ([18F]FDOPA) (n = 3), or [68Ga] labelled quinoline-based small molecule fibroblast activation protein inhibitor ([68Ga]FAPI-46) (n = 4) were injected intravenously if solid tumours were detected with MRI. For time-activity curves for [18F]FET, additional micro PET (µPET) was performed. The chick embryos were sacrificed 60 min post-injection, and cryosections of the tumour-bearing brains were produced and evaluated with autoradiography and immunohistochemistry. RESULTS Intracerebral tumours were produced with a 100% success rate in viable chick embryos at the experimental endpoint. However, 52% of chick embryos (n = 85) did not survive the procedure to embryonic development day 20. For the evaluated radiotracers, the tumour-to-brain ratios (TBR) derived from ex vivo autoradiography, as well as the tracer kinetics derived from µPET for intracerebral chick embryo tumours, were comparable to those previously reported in rodents and patients: the TBRmean for [18F]FET was 1.69 ± 0.54 (n = 5), and 3.8 for one hypermetabolic tumour and < 2.0 for two isometabolic tumors using [18F]FDOPA, with a TBRmean of 1.92 ± 1,11 (n = 3). The TBRmean of [68Ga]FAPI-46 for intracerebral chick embryo tumours was 19.13 ± 0.64 (n = 4). An intact blood-tumour barrier was observed in one U87-MG tumour (n = 5). CONCLUSIONS Radiotracer imaging of intracerebral tumours in the chick embryo offers a fast model for the evaluation of radiotracer uptake, accumulation, and kinetics. Our results indicate a high comparability between intracerebral tumour imaging in chick embryos and xenograft rodent models or brain tumour patients.
Collapse
Affiliation(s)
- Sandra Krause
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany.
| | - Alexandru Florea
- Department of Nuclear Medicine, RWTH Aachen University Hospital, Aachen, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Chang-Hoon Choi
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Wieland A Worthoff
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Alexander Heinzel
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
- Department for Nuclear Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Saskia Fischer
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Nicole Burda
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Bernd Neumaier
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - N Jon Shah
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
- Department of Neurology, RWTH Aachen University Hospital, Aachen, Germany
- JARA - BRAIN - Translational Medicine, Aachen, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
- Department of Nuclear Medicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, RWTH Aachen University Hospital, Aachen, Germany
- JARA - BRAIN - Translational Medicine, Aachen, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
- Department of Nuclear Medicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Carina Stegmayr
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
- Department of Nuclear Medicine, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
2
|
Hou L, Chen Z, Chen F, Sheng L, Ye W, Dai Y, Guo X, Dong C, Li G, Liao K, Li Y, Ma J, Wei H, Ran W, Shang J, Ling X, Patel JS, Liang SH, Xu H, Wang L. Synthesis, preclinical assessment, and first-in-human study of [ 18F]d 4-FET for brain tumor imaging. Eur J Nucl Med Mol Imaging 2025; 52:864-875. [PMID: 39482500 DOI: 10.1007/s00259-024-06964-8] [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: 07/02/2024] [Accepted: 10/18/2024] [Indexed: 11/03/2024]
Abstract
PURPOSE Tumor-to-background ratio (TBR) is a critical metric in oncologic PET imaging. This study aims to enhance the TBR of [18F]FET in brain tumor imaging by substituting deuterium ("D") for hydrogen ("H"), thereby improving the diagnostic sensitivity and accuracy. METHODS [18F]d4-FET was synthesised by two automated radiochemistry modules. Biodistribution studies and imaging efficacy were evaluated in vivo and ex vivo in rodent models, while metabolic stability and radiation dosimetry were assessed in non-human primates. Additionally, preliminary imaging evaluations were carried out in five brain tumor patients: three glioma patients underwent imaging with both [18F]d4-FET and [18F]FET, and two patients with brain metastases were imaged using [18F]d4-FET and [18F]FDG. RESULTS [18F]d4-FET demonstrated high radiochemical purity and yield. PET/MRI in rodent models demonstrated superior TBR for [18F]d4-FET compared to [18F]FET, and autoradiography showed tumor margins that correlated well with pathological extents. Studies in cynomolgus monkeys indicated comparable in vivo stability and effective dose with [18F]FET. In glioma patients, [18F]d4-FET showed enhanced TBR, while in patients with brain metastases, [18F]d4-FET displayed superior lesion delineation compared to [18F]FDG, especially in smaller metastatic sites. CONCLUSION We successfully synthesized the novel PET radiotracer [18F]d4-FET, which retains the advantageous properties of [18F]FET while potentially enhancing TBR for glioma imaging. Preliminary studies indicate excellent stability, efficacy, and sensitivity of [18F]d4-FET, suggesting its potential in clinical evaluations of brain tumors. TRIAL REGISTRATION ChiCTR2400081576, registration date: 2024-03-05, https://www.chictr.org.cn/bin/project/edit?pid=206162.
Collapse
Affiliation(s)
- Lu Hou
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Zhiyong Chen
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Fanfan Chen
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518025, China
| | - Lianghe Sheng
- Department of Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Weijian Ye
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Yingchu Dai
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Xiaoyu Guo
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Chenchen Dong
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Guocong Li
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Kai Liao
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Yinlong Li
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA, 30322, USA
| | - Jie Ma
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Huiyi Wei
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Wenqing Ran
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jingjie Shang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Xueying Ling
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jimmy S Patel
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA, 30322, USA
| | - Steven H Liang
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA, 30322, USA.
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
| |
Collapse
|
3
|
Kaizuka Y, Suzuki H, Watabe T, Ooe K, Toyoshima A, Takahashi K, Sawada K, Iimori T, Masuda Y, Uno T, Kannaka K, Uehara T. Neopentyl glycol-based radiohalogen-labeled amino acid derivatives for cancer radiotheranostics. EJNMMI Radiopharm Chem 2024; 9:17. [PMID: 38407647 PMCID: PMC10897087 DOI: 10.1186/s41181-024-00244-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/13/2024] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND L-type amino acid transporter 1 (LAT1) is overexpressed in various cancers; therefore, radiohalogen-labeled amino acid derivatives targeting LAT1 have emerged as promising candidates for cancer radiotheranostics. However, 211At-labeled amino acid derivatives exhibit instability against deastatination in vivo, making it challenging to use 211At for radiotherapy. In this study, radiohalogen-labeled amino acid derivatives with high dehalogenation stability were developed. RESULTS We designed and synthesized new radiohalogen-labeled amino acid derivatives ([211At]At-NpGT, [125I]I-NpGT, and [18F]F-NpGT) in which L-tyrosine was introduced into the neopentyl glycol (NpG) structure. The radiolabeled amino acid derivatives were recognized as substrates of LAT1 in the in vitro studies using C6 glioma cells. In a biodistribution study using C6 glioma-bearing mice, these agents exhibited high stability against in vivo dehalogenation and similar biodistributions. The similarity of [211At]At-NpGT and [18F]F-NpGT indicated that these pairs of radiolabeled compounds would be helpful in radiotheranostics. Moreover, [211At]At-NpGT exhibited a dose-dependent inhibitory effect on the growth of C6 glioma-bearing mice. CONCLUSIONS [211At]At-NpGT exhibited a dose-dependent inhibitory effect on the tumor growth of glioma-bearing mice, and its biodistribution was similar to that of other radiohalogen-labeled amino acid derivatives. These findings suggest that radiotheranostics using [18F]F-NpGT and [123/131I]I-NpGT for diagnostic applications and [211At]At-NpGT and [131I]I-NpGT for therapeutic applications are promising.
Collapse
Affiliation(s)
- Yuta Kaizuka
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8675, Japan
| | - Hiroyuki Suzuki
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8675, Japan.
| | - Tadashi Watabe
- Department of Nuclear Medicine and Tracer Kinetics, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Institute for Radiation Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Kazuhiro Ooe
- Institute for Radiation Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Atsushi Toyoshima
- Institute for Radiation Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Kazuhiro Takahashi
- Advanced Clinical Research Center, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Koichi Sawada
- Department of Radiology, Chiba University Hospital, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8677, Japan
| | - Takashi Iimori
- Department of Radiology, Chiba University Hospital, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8677, Japan
| | - Yoshitada Masuda
- Department of Radiology, Chiba University Hospital, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8677, Japan
| | - Takashi Uno
- Department of Radiology, Chiba University Hospital, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8677, Japan
- Diagnostic Radiology and Radiation Oncology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8670, Japan
| | - Kento Kannaka
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8675, Japan
| | - Tomoya Uehara
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba, 260-8675, Japan.
| |
Collapse
|
4
|
Two Decades of Brain Tumour Imaging with O-(2-[18F]fluoroethyl)-L-tyrosine PET: The Forschungszentrum Jülich Experience. Cancers (Basel) 2022; 14:cancers14143336. [PMID: 35884396 PMCID: PMC9319157 DOI: 10.3390/cancers14143336] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/28/2022] [Accepted: 07/05/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary PET using radiolabelled amino acids has become an essential tool for diagnosing brain tumours in addition to MRI. O-(2-[18F]fluoroethyl)-L-tyrosine (FET) is one of the most successful tracers in the field. We analysed our database of 6534 FET PET examinations regarding the diagnostic needs and preferences of the referring physicians for FET PET in the clinical decision-making process. The demand for FET PET increased considerably in the last decade, especially for differentiating tumour progress from treatment-related changes in gliomas. Accordingly, referring physicians rated the diagnostics of recurrent glioma and recurrent brain metastases as the most relevant indication for FET PET. The analysis and survey results confirm the high relevance of FET PET in the clinical diagnosis of brain tumours and support the need for approval for routine use. Abstract O-(2-[18F]fluoroethyl)-L-tyrosine (FET) is a widely used amino acid tracer for positron emission tomography (PET) imaging of brain tumours. This retrospective study and survey aimed to analyse our extensive database regarding the development of FET PET investigations, indications, and the referring physicians’ rating concerning the role of FET PET in the clinical decision-making process. Between 2006 and 2019, we performed 6534 FET PET scans on 3928 different patients against a backdrop of growing demand for FET PET. In 2019, indications for the use of FET PET were as follows: suspected recurrent glioma (46%), unclear brain lesions (20%), treatment monitoring (19%), and suspected recurrent brain metastasis (13%). The referring physicians were neurosurgeons (60%), neurologists (19%), radiation oncologists (11%), general oncologists (3%), and other physicians (7%). Most patients travelled 50 to 75 km, but 9% travelled more than 200 km. The role of FET PET in decision-making in clinical practice was evaluated by a questionnaire consisting of 30 questions, which was filled out by 23 referring physicians with long experience in FET PET. Fifty to seventy per cent rated FET PET as being important for different aspects of the assessment of newly diagnosed gliomas, including differential diagnosis, delineation of tumour extent for biopsy guidance, and treatment planning such as surgery or radiotherapy, 95% for the diagnosis of recurrent glioma, and 68% for the diagnosis of recurrent brain metastases. Approximately 50% of the referring physicians rated FET PET as necessary for treatment monitoring in patients with glioma or brain metastases. All referring physicians stated that the availability of FET PET is essential and that it should be approved for routine use. Although the present analysis is limited by the fact that only physicians who frequently referred patients for FET PET participated in the survey, the results confirm the high relevance of FET PET in the clinical diagnosis of brain tumours and support the need for its approval for routine use.
Collapse
|
5
|
Cohen AS, Grudzinski J, Smith GT, Peterson TE, Whisenant JG, Hickman TL, Ciombor KK, Cardin D, Eng C, Goff LW, Das S, Coffey RJ, Berlin JD, Manning HC. First-in-Human PET Imaging and Estimated Radiation Dosimetry of l-[5- 11C]-Glutamine in Patients with Metastatic Colorectal Cancer. J Nucl Med 2022; 63:36-43. [PMID: 33931465 PMCID: PMC8717201 DOI: 10.2967/jnumed.120.261594] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/26/2021] [Indexed: 12/23/2022] Open
Abstract
Altered metabolism is a hallmark of cancer. In addition to glucose, glutamine is an important nutrient for cellular growth and proliferation. Noninvasive imaging via PET may help facilitate precision treatment of cancer through patient selection and monitoring of treatment response. l-[5-11C]-glutamine (11C-glutamine) is a PET tracer designed to study glutamine uptake and metabolism. The aim of this first-in-human study was to evaluate the radiologic safety and biodistribution of 11C-glutamine for oncologic PET imaging. Methods: Nine patients with confirmed metastatic colorectal cancer underwent PET/CT imaging. Patients received 337.97 ± 44.08 MBq of 11C-glutamine. Dynamic PET acquisitions that were centered over the abdomen or thorax were initiated simultaneously with intravenous tracer administration. After the dynamic acquisition, a whole-body PET/CT scan was acquired. Volume-of-interest analyses were performed to obtain estimates of organ-based absorbed doses of radiation. Results:11C-glutamine was well tolerated in all patients, with no observed safety concerns. The organs with the highest radiation exposure included the bladder, pancreas, and liver. The estimated effective dose was 4.46E-03 ± 7.67E-04 mSv/MBq. Accumulation of 11C-glutamine was elevated and visualized in lung, brain, bone, and liver metastases, suggesting utility for cancer imaging. Conclusion: PET using 11C-glutamine appears safe for human use and allows noninvasive visualization of metastatic colon cancer lesions in multiple organs. Further studies are needed to elucidate its potential for other cancers and for monitoring response to treatment.
Collapse
Affiliation(s)
- Allison S Cohen
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Gary T Smith
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Section Chief, Nuclear Medicine, Tennessee Valley Healthcare System, Nashville VA Medical Center, Nashville, Tennessee
| | - Todd E Peterson
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jennifer G Whisenant
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Tiffany L Hickman
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Kristen K Ciombor
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Dana Cardin
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Cathy Eng
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Laura W Goff
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Satya Das
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Robert J Coffey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee; and
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| | - Jordan D Berlin
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - H Charles Manning
- Vanderbilt Center for Molecular Probes, Vanderbilt University Medical Center, Nashville, Tennessee;
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee; and
| |
Collapse
|
6
|
Beinat C, Patel CB, Haywood T, Murty S, Naya L, Castillo JB, Reyes ST, Phillips M, Buccino P, Shen B, Park JH, Koran MEI, Alam IS, James ML, Holley D, Halbert K, Gandhi H, He JQ, Granucci M, Johnson E, Liu DD, Uchida N, Sinha R, Chu P, Born DE, Warnock GI, Weissman I, Hayden-Gephart M, Khalighi M, Massoud TF, Iagaru A, Davidzon G, Thomas R, Nagpal S, Recht LD, Gambhir SS. A Clinical PET Imaging Tracer ([ 18F]DASA-23) to Monitor Pyruvate Kinase M2-Induced Glycolytic Reprogramming in Glioblastoma. Clin Cancer Res 2021; 27:6467-6478. [PMID: 34475101 PMCID: PMC8639752 DOI: 10.1158/1078-0432.ccr-21-0544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 07/15/2021] [Accepted: 08/30/2021] [Indexed: 01/10/2023]
Abstract
PURPOSE Pyruvate kinase M2 (PKM2) catalyzes the final step in glycolysis, a key process of cancer metabolism. PKM2 is preferentially expressed by glioblastoma (GBM) cells with minimal expression in healthy brain. We describe the development, validation, and translation of a novel PET tracer to study PKM2 in GBM. We evaluated 1-((2-fluoro-6-[18F]fluorophenyl)sulfonyl)-4-((4-methoxyphenyl)sulfonyl)piperazine ([18F]DASA-23) in cell culture, mouse models of GBM, healthy human volunteers, and patients with GBM. EXPERIMENTAL DESIGN [18F]DASA-23 was synthesized with a molar activity of 100.47 ± 29.58 GBq/μmol and radiochemical purity >95%. We performed initial testing of [18F]DASA-23 in GBM cell culture and human GBM xenografts implanted orthotopically into mice. Next, we produced [18F]DASA-23 under FDA oversight, and evaluated it in healthy volunteers and a pilot cohort of patients with glioma. RESULTS In mouse imaging studies, [18F]DASA-23 clearly delineated the U87 GBM from surrounding healthy brain tissue and had a tumor-to-brain ratio of 3.6 ± 0.5. In human volunteers, [18F]DASA-23 crossed the intact blood-brain barrier and was rapidly cleared. In patients with GBM, [18F]DASA-23 successfully outlined tumors visible on contrast-enhanced MRI. The uptake of [18F]DASA-23 was markedly elevated in GBMs compared with normal brain, and it identified a metabolic nonresponder within 1 week of treatment initiation. CONCLUSIONS We developed and translated [18F]DASA-23 as a new tracer that demonstrated the visualization of aberrantly expressed PKM2 for the first time in human subjects. These results warrant further clinical evaluation of [18F]DASA-23 to assess its utility for imaging therapy-induced normalization of aberrant cancer metabolism.
Collapse
Affiliation(s)
- Corinne Beinat
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California.
| | - Chirag B Patel
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Tom Haywood
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Surya Murty
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Lewis Naya
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Jessa B Castillo
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Samantha T Reyes
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Megan Phillips
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Pablo Buccino
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Bin Shen
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Jun Hyung Park
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Mary Ellen I Koran
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Israt S Alam
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Michelle L James
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Dawn Holley
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Kim Halbert
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Harsh Gandhi
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Joy Q He
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Monica Granucci
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Eli Johnson
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Daniel Dan Liu
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Nobuko Uchida
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Rahul Sinha
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Pauline Chu
- Stanford Human Research Histology Core, Stanford University School of Medicine, Stanford, California
| | - Donald E Born
- Department of Pathology, Neuropathology, Stanford University School of Medicine, Stanford, California
| | | | - Irving Weissman
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Melanie Hayden-Gephart
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Mehdi Khalighi
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Tarik F Massoud
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
- Division of Neuroimaging and Neurointervention, Department of Radiology, Stanford University School of Medicine, Stanford, California
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Guido Davidzon
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Reena Thomas
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Seema Nagpal
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Lawrence D Recht
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California.
| | - Sanjiv Sam Gambhir
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
- Departments of Bioengineering and Materials Science & Engineering, Stanford University, Stanford, California
| |
Collapse
|
7
|
Moon H, Byun BH, Lim I, Kim BI, Choi CW, Rhee CH, Lee KC, Woo SK, Park C, Kil HS, Chi DY, Youn SM, Lim SM. A Phase 0 Microdosing PET/CT Study Using O-[18F]Fluoromethyl-d-Tyrosine in Normal Human Brain and Brain Tumor. Clin Nucl Med 2021; 46:717-722. [PMID: 34034333 DOI: 10.1097/rlu.0000000000003735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE The aim of the present study was to obtain information about distribution, radiation dosimetry, toxicity, and pharmacokinetics of O-[18F]fluoromethyl-d-tyrosine (d-18F-FMT), an amino acid PET tracer, in patients with brain tumors. PATIENTS AND METHODS A total of 6 healthy controls (age = 19-25 years, 3 males and 3 females) with brain PET images and radiation dosimetry and 12 patients (median age = 60 years, 6 males and 6 females) with primary (n = 5) or metastatic brain tumor (n = 7) were enrolled. We acquired 60-minute dynamic brain PET images after injecting 370 MBq of d-18F-FMT. Time-activity curves of d-18F-FMT uptake in normal brain versus brain tumors and tumor-to-background ratio were analyzed for each PET data set. RESULTS Normal cerebral uptake of d-18F-FMT decreased from 0 to 5 minutes after injection, but gradually increased from 10 to 60 minutes. Tumoral uptake of d-18F-FMT reached a peak before 30 minutes. Tumor-to-background ratio peaked at less than 15 minutes for 8 patients and more than 15 minutes for 4 patients. The mean effective dose was calculated to be 13.2 μSv/MBq. CONCLUSIONS Using d-18F-FMT as a PET radiotracer is safe. It can distinguish brain tumor from surrounding normal brain tissues with a high contrast. Early-time PET images of brain tumors should be acquired because the tumor-to-background ratio tended to reach a peak within 15 minutes after injection.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kyo Chul Lee
- Division of RI Convergence, Korea Institute of Radiological and Medical Sciences
| | - Sang-Keun Woo
- Division of RI Convergence, Korea Institute of Radiological and Medical Sciences
| | | | | | | | | | | |
Collapse
|
8
|
Kanai Y. Amino acid transporter LAT1 (SLC7A5) as a molecular target for cancer diagnosis and therapeutics. Pharmacol Ther 2021; 230:107964. [PMID: 34390745 DOI: 10.1016/j.pharmthera.2021.107964] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 07/30/2021] [Indexed: 01/13/2023]
Abstract
Cancer cells require a massive supply of nutrients, including sugars and amino acids-the upregulation of transporters for each nutrient contributes to meet the demand. Distinct from glucose transporters, amino acid transporters include ones whose expression is specific to cancer cells. For example, LAT1 (SLC7A5) displays protein expression mostly limited to the plasma membrane of cancer cells. The exceptions are the placental barrier and the blood-brain barrier, where immunohistochemical and mass spectrometric studies have shown LAT1 expression, although their levels are supposed to be lower than those in cancers. The expression of LAT1 has been reported in cancers from various tissue origins, where high LAT1 expression is related to the poor prognosis of patients. LAT1 is essential for cancer cell growth because the pharmacologic inhibition and knockdown/knockout of LAT1 suppress the proliferation of cancer cells and the growth of xenograft tumors. The inhibition of LAT1 suppresses protein synthesis by downregulating the mTORC1 signaling pathway and mobilizing the general amino acid control (GAAC) pathway in cancer cells. LAT1 is, thus, a candidate molecular target for the diagnosis and therapeutics of cancers. 18F-labeled 3-fluoro-l-α-methyl-tyrosine (FAMT) is used as a LAT1-specific PET probe for cancer detection due to the LAT1 specificity of α-methyl aromatic amino acids. FAMT accumulation is cancer-specific and avoids non-cancer lesions, including inflammation, confirming the cancer-specific expression of LAT1 in humans. Due to the cancer-specific nature, LAT1 can also be used for cancer-specific delivery of anti-tumor agents such as l-para-boronophenylalanine used for boron neutron capture therapy and α-emitting nuclide-labeled LAT1 substrates developed for nuclear medicine treatment. Based on the importance of LAT1 in cancer progression, high-affinity LAT1-specific inhibitors have been developed for anti-tumor drugs. JPH203 (KYT0353) is such a compound designed based on the structure-activity relationship of LAT1 ligands. It is one of the highest-affinity inhibitors with less affecting other transporters. It suppresses tumor growth in vivo without significant toxicity in preclinical studies at doses enough to suppress tumor growth. In the phase-I clinical trial, JPH203 appeared to provide promising activity. Because the mechanisms of action of LAT1 inhibitors are novel, with or without combination with other anti-tumor drugs, they could contribute to the treatment of cancers that do not respond to current therapy. The LAT1-specific PET probe could also be used as companion diagnostics of the LAT1-targeting therapies to select patients to whom therapeutic benefits could be expected. Recently, the cryo-EM structure of LAT1 has been solved, which would facilitate the understanding of the mechanisms of the dynamic interaction of ligands and the binding site, and further designing new compounds with higher activity.
Collapse
Affiliation(s)
- Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
9
|
Furtak J, Rakowska J, Szylberg T, Harat M, Małkowski B, Harat M. Glioma Biopsy Based on Hybrid Dual Time-Point FET-PET/MRI-A Proof of Concept Study. Front Neurol 2021; 12:634609. [PMID: 34046002 PMCID: PMC8144440 DOI: 10.3389/fneur.2021.634609] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 03/15/2021] [Indexed: 11/13/2022] Open
Abstract
Neuroimaging based on O-[2-(18F)fluoroethyl]-l-tyrosine (FET)-PET provides additional information on tumor grade and extent compared with MRI. Dynamic PET for biopsy target selection further improves results but is often clinically impractical. Static FET-PET performed at two time-points may be a good compromise, but data on this approach are limited. The aim of this study was to compare the histology of lesions obtained from two challenging glioma patients with targets selected based on hybrid dual time-point FET-PET/MRI. Five neuronavigated tumor biopsies were performed in two difficult cases of suspected glioma. Lesions with (T1-CE) and without contrast enhancement (T1 and T2-FLAIR) on MRI were selected. Dual time-point FET-PET imaging was performed 5–15 min (PET10) and 45–60 min (PET60) after radionuclide injection. The most informative FET-PET/MRI images were coregistered with MRI in time of biopsy planning. Five biopsy targets (three from high uptake and two from moderate uptake FET areas) thought to represent the most malignant sites and tumor extent were selected. Histopathological findings were compared with FET-PET and MRI images. Increased FET uptake in the area of non-CE locations on MRI correlated well with high-grade gliomas localized as far as 3 cm from T1-CE foci. Selecting a target in the motor cortex based on FET kinetics defined by dual time-point PET resulted in a grade IV diagnosis after previous negative biopsies based on MRI. An additional grade III diagnosis was obtained from an area of glioma infiltration with moderate FET uptake (between 1 and 1.25 SUV). These findings seem to show that dual time-point FET-PET-based biopsies can provide additional and clinically useful information for glioma diagnosis. Selection of targets based on dual time-point images may be useful for determining the most malignant tumor areas and may therefore be useful for resection and radiotherapy planning.
Collapse
Affiliation(s)
- Jacek Furtak
- Department of Neurosurgery, 10th Military Research Hospital, Bydgoszcz, Poland
| | - Józefina Rakowska
- Department of Neurosurgery, 10th Military Research Hospital, Bydgoszcz, Poland
| | - Tadeusz Szylberg
- Department of Pathomorphology, 10th Military Research Hospital, Bydgoszcz, Poland
| | - Marek Harat
- Department of Neurosurgery, 10th Military Research Hospital, Bydgoszcz, Poland.,Department of Neurosurgery and Neurology, Faculty of Health Sciences, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Bogdan Małkowski
- Department of Positron Emission Tomography and Molecular Imaging, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland.,Department of Nuclear Medicine, Franciszek Lukaszczyk Oncology Center, Bydgoszcz, Poland
| | - Maciej Harat
- Department of Oncology and Brachytherapy, Faculty of Medicine, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland.,Department of Neurooncology and Radiosurgery, Franciszek Lukaszczyk Oncology Center, Bydgoszcz, Poland
| |
Collapse
|
10
|
Kebir S, Rauschenbach L, Weber M, Lazaridis L, Schmidt T, Keyvani K, Schäfer N, Milia A, Umutlu L, Pierscianek D, Stuschke M, Forsting M, Sure U, Kleinschnitz C, Antoch G, Colletti PM, Rubello D, Herrmann K, Herrlinger U, Scheffler B, Bundschuh RA, Glas M. Machine learning-based differentiation between multiple sclerosis and glioma WHO II°-IV° using O-(2-[18F] fluoroethyl)-L-tyrosine positron emission tomography. J Neurooncol 2021; 152:325-332. [PMID: 33502678 DOI: 10.1007/s11060-021-03701-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/13/2021] [Indexed: 11/28/2022]
Abstract
INTRODUCTION This study aimed to test the diagnostic significance of FET-PET imaging combined with machine learning for the differentiation between multiple sclerosis (MS) and glioma II°-IV°. METHODS Our database was screened for patients in whom FET-PET imaging was performed for the diagnostic workup of newly diagnosed lesions evident on MRI and suggestive of glioma. Among those, we identified patients with histologically confirmed glioma II°-IV°, and those who later turned out to have MS. For each group, tumor-to-brain ratio (TBR) derived features of FET were determined. A support vector machine (SVM) based machine learning algorithm was constructed to enhance classification ability, and Receiver Operating Characteristic (ROC) analysis with area under the curve (AUC) metric served to ascertain model performance. RESULTS A total of 41 patients met selection criteria, including seven patients with MS and 34 patients with glioma. TBR values were significantly higher in the glioma group (TBRmax glioma vs. MS: p = 0.002; TBRmean glioma vs. MS: p = 0.014). In a subgroup analysis, TBR values significantly differentiated between MS and glioblastoma (TBRmax glioblastoma vs. MS: p = 0.0003, TBRmean glioblastoma vs. MS: p = 0.0003) and between MS and oligodendroglioma (ODG) (TBRmax ODG vs. MS: p = 0.003; TBRmean ODG vs. MS: p = 0.01). The ability to differentiate between MS and glioma II°-IV° increased from 0.79 using standard TBR analysis to 0.94 using a SVM based machine learning algorithm. CONCLUSIONS FET-PET imaging may help differentiate MS from glioma II°-IV° and SVM based machine learning approaches can enhance classification performance.
Collapse
Affiliation(s)
- Sied Kebir
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.,West German Cancer Center (WTZ), German Cancer Consortium (DKTK), University Hospital Essen, University Duisburg-Essen, Partner Site University Hospital Essen, Essen, Germany.,DKFZ Division of Translational Neurooncology at the West German Cancer Center (WTZ), German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Laurèl Rauschenbach
- DKFZ Division of Translational Neurooncology at the West German Cancer Center (WTZ), German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany.,Department of Neurosurgery and Spine Surgery, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Manuel Weber
- Department of Nuclear Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Lazaros Lazaridis
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.,West German Cancer Center (WTZ), German Cancer Consortium (DKTK), University Hospital Essen, University Duisburg-Essen, Partner Site University Hospital Essen, Essen, Germany.,DKFZ Division of Translational Neurooncology at the West German Cancer Center (WTZ), German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Teresa Schmidt
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.,West German Cancer Center (WTZ), German Cancer Consortium (DKTK), University Hospital Essen, University Duisburg-Essen, Partner Site University Hospital Essen, Essen, Germany
| | - Kathy Keyvani
- Institute of Neuropathology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Niklas Schäfer
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Asma Milia
- Department of Pulmonology and Cardiology, Petrus Hospital Academic Teaching, Wuppertal, Germany
| | - Lale Umutlu
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Daniela Pierscianek
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Martin Stuschke
- Department of Radiotherapy, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Michael Forsting
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ulrich Sure
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Christoph Kleinschnitz
- Department of Neurology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Gerald Antoch
- Department of Diagnostic and Interventional Radiology, University Hospital Düsseldorf, University of Düsseldorf, Düsseldorf, Germany
| | - Patrick M Colletti
- Department of Radiology, University of Southern California, Los Angeles, USA
| | - Domenico Rubello
- Department of Nuclear Medicine, Radiology, Neuroradiology, Clinical Pathology, S. Maria Della Misericordia Hospital, Rovigo, Italy
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ulrich Herrlinger
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Björn Scheffler
- West German Cancer Center (WTZ), German Cancer Consortium (DKTK), University Hospital Essen, University Duisburg-Essen, Partner Site University Hospital Essen, Essen, Germany.,DKFZ Division of Translational Neurooncology at the West German Cancer Center (WTZ), German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Ralph A Bundschuh
- Department of Nuclear Medicine, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Martin Glas
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany. .,West German Cancer Center (WTZ), German Cancer Consortium (DKTK), University Hospital Essen, University Duisburg-Essen, Partner Site University Hospital Essen, Essen, Germany. .,DKFZ Division of Translational Neurooncology at the West German Cancer Center (WTZ), German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany. .,Division of Clinical Neurooncology, Department of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany.
| |
Collapse
|
11
|
Stegmayr C, Stoffels G, Filß C, Heinzel A, Lohmann P, Willuweit A, Ermert J, Coenen HH, Mottaghy FM, Galldiks N, Langen KJ. Current trends in the use of O-(2-[ 18F]fluoroethyl)-L-tyrosine ([ 18F]FET) in neurooncology. Nucl Med Biol 2021; 92:78-84. [PMID: 32113820 DOI: 10.1016/j.nucmedbio.2020.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 02/16/2020] [Indexed: 12/14/2022]
Abstract
The diagnostic potential of PET using the amino acid analogue O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) in brain tumor diagnostics has been proven in many studies during the last two decades and is still the subject of multiple studies every year. In addition to standard magnetic resonance imaging (MRI), positron emission tomography (PET) using [18F]FET provides important diagnostic data concerning brain tumor delineation, therapy planning, treatment monitoring, and improved differentiation between treatment-related changes and tumor recurrence. The pharmacokinetics, uptake mechanisms and metabolism have been well described in various preclinical studies. The accumulation of [18F]FET in most benign lesions and healthy brain tissue has been shown to be low, thus providing a high contrast between tumor tissue and benign tissue alterations. Based on logistic advantages of F-18 labelling and convincing clinical results, [18F]FET has widely replaced short lived amino acid tracers such as L-[11C]methyl-methionine ([11C]MET) in many centers across Western Europe. This review summarizes the basic knowledge on [18F]FET and its contribution to the care of patients with brain tumors. In particular, recent studies about specificity, possible pitfalls, and the utility of [18F]FET PET in tumor grading and prognostication regarding the revised WHO classification of brain tumors are addressed.
Collapse
Affiliation(s)
- Carina Stegmayr
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany
| | - Christian Filß
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany; Dept. of Nuclear Medicine, RWTH University Hospital, Aachen, Germany
| | - Alexander Heinzel
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany; Dept. of Nuclear Medicine, RWTH University Hospital, Aachen, Germany; Juelich-Aachen Research Alliance (JARA) - Section JARA-Brain, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany
| | - Antje Willuweit
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany
| | - Johannes Ermert
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany
| | - Heinz H Coenen
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany
| | - Felix M Mottaghy
- Dept. of Nuclear Medicine, RWTH University Hospital, Aachen, Germany; Juelich-Aachen Research Alliance (JARA) - Section JARA-Brain, Germany; Center of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Duesseldorf, Germany; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, the Netherlands
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany; Dept. of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Duesseldorf, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, INM-4, INM-5), Forschungszentrum Juelich, Juelich, Germany; Dept. of Nuclear Medicine, RWTH University Hospital, Aachen, Germany; Juelich-Aachen Research Alliance (JARA) - Section JARA-Brain, Germany; Center of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Duesseldorf, Germany.
| |
Collapse
|
12
|
Snyder SE, Butch ER, Shulkin BL. Radiopharmaceuticals in Pediatric Nuclear Medicine. HANDBOOK OF RADIOPHARMACEUTICALS 2020:653-701. [DOI: 10.1002/9781119500575.ch21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
13
|
Giesel FL, Adeberg S, Syed M, Lindner T, Jiménez-Franco LD, Mavriopoulou E, Staudinger F, Tonndorf-Martini E, Regnery S, Rieken S, El Shafie R, Röhrich M, Flechsig P, Kluge A, Altmann A, Debus J, Haberkorn U, Kratochwil C. FAPI-74 PET/CT Using Either 18F-AlF or Cold-Kit 68Ga Labeling: Biodistribution, Radiation Dosimetry, and Tumor Delineation in Lung Cancer Patients. J Nucl Med 2020; 62:201-207. [PMID: 32591493 PMCID: PMC8679591 DOI: 10.2967/jnumed.120.245084] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/27/2020] [Indexed: 12/14/2022] Open
Abstract
68Ga-fibroblast activation protein inhibitors (FAPIs) 2, 4, and 46 have already been proposed as promising PET tracers. However, the short half-life of 68Ga (68 min) creates problems with manufacture and delivery. 18F (half-life, 110 min) labeling would result in a more practical large-scale production, and a cold-kit formulation would improve the spontaneous availability. The NOTA chelator ligand FAPI-74 can be labeled with both 18F-AlF and 68Ga. Here, we describe the in vivo evaluation of 18F-FAPI-74 and a proof of mechanism for 68Ga-FAPI-74 labeled at ambient temperature. Methods: In 10 patients with lung cancer, PET scans were acquired at 10 min, 1 h, and 3 h after administration of 259 ± 26 MBq of 18F-FAPI-74. Physiologic biodistribution and tumor uptake were semiquantitatively evaluated on the basis of SUV at each time point. Absorbed doses were evaluated using OLINDA/EXM, version 1.1, and QDOSE dosimetry software with the dose calculator IDAC-Dose, version 2.1. Identical methods were used to evaluate one examination after injection of 263 MBq of 68Ga-FAPI-74. Results: The highest contrast was achieved in primary tumors, lymph nodes, and distant metastases at 1 h after injection, with an SUVmax of more than 10. The effective dose per a 100-MBq administered activity of 18F-FAPI-74 was 1.4 ± 0.2 mSv, and for 68Ga-FAPI-74 it was 1.6 mSv. Thus, the radiation burden of a diagnostic 18F-FAPI-74 PET scan is even lower than that of PET scans with 18F-FDG and other 18F tracers; 68Ga-FAPI-74 is comparable to other 68Ga ligands. FAPI PET/CT supported target volume definition for guiding radiotherapy. Conclusion: The high contrast and low radiation burden of FAPI-74 PET/CT favor multiple clinical applications. Centralized large-scale production of 18F-FAPI-74 or decentralized cold-kit labeling of 68Ga-FAPI-74 allows flexible routine use.
Collapse
Affiliation(s)
- Frederik L Giesel
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Sebastian Adeberg
- Heidelberg Institute of Radiation Oncology, Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center, Heidelberg, Germany.,Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center, Heidelberg, Germany
| | - Mustafa Syed
- Heidelberg Institute of Radiation Oncology, Heidelberg, Germany.,Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Lindner
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Eleni Mavriopoulou
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Fabian Staudinger
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Eric Tonndorf-Martini
- Heidelberg Institute of Radiation Oncology, Heidelberg, Germany.,Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Sebastian Regnery
- Heidelberg Institute of Radiation Oncology, Heidelberg, Germany.,Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Rieken
- Heidelberg Institute of Radiation Oncology, Heidelberg, Germany.,Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center, Heidelberg, Germany.,Department of Radiation Oncology, University Hospital Göttingen, Göttingen, Germany; and
| | - Rami El Shafie
- Heidelberg Institute of Radiation Oncology, Heidelberg, Germany.,Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center, Heidelberg, Germany
| | - Manuel Röhrich
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Paul Flechsig
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Andreas Kluge
- ABX-CRO Advanced Pharmaceutical Services Forschungsgesellschaft mbH, Dresden, Germany
| | - Annette Altmann
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Jürgen Debus
- Heidelberg Institute of Radiation Oncology, Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center, Heidelberg, Germany.,Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center, Heidelberg, Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany .,Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center, Heidelberg, Germany
| | - Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
14
|
Allott L, Aboagye EO. Chemistry Considerations for the Clinical Translation of Oncology PET Radiopharmaceuticals. Mol Pharm 2020; 17:2245-2259. [DOI: 10.1021/acs.molpharmaceut.0c00328] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Louis Allott
- Comprehensive Cancer Imaging Centre, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, United Kingdom
| | - Eric O. Aboagye
- Comprehensive Cancer Imaging Centre, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, United Kingdom
| |
Collapse
|
15
|
Stegmayr C, Willuweit A, Lohmann P, Langen KJ. O-(2-[18F]-Fluoroethyl)-L-Tyrosine (FET) in Neurooncology: A Review of Experimental Results. Curr Radiopharm 2020; 12:201-210. [PMID: 30636621 DOI: 10.2174/1874471012666190111111046] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 11/22/2022]
Abstract
In recent years, PET using radiolabelled amino acids has gained considerable interest as an additional tool besides MRI to improve the diagnosis of cerebral gliomas and brain metastases. A very successful tracer in this field is O-(2-[18F]fluoroethyl)-L-tyrosine (FET) which in recent years has replaced short-lived tracers such as [11C]-methyl-L-methionine in many neuro-oncological centers in Western Europe. FET can be produced with high efficiency and distributed in a satellite concept like 2- [18F]fluoro-2-deoxy-D-glucose. Many clinical studies have demonstrated that FET PET provides important diagnostic information regarding the delineation of cerebral gliomas for therapy planning, an improved differentiation of tumor recurrence from treatment-related changes and sensitive treatment monitoring. In parallel, a considerable number of experimental studies have investigated the uptake mechanisms of FET on the cellular level and the behavior of the tracer in various benign lesions in order to clarify the specificity of FET uptake for tumor tissue. Further studies have explored the effects of treatment related tissue alterations on tracer uptake such as surgery, radiation and drug therapy. Finally, the role of blood-brain barrier integrity for FET uptake which presents an important aspect for PET tracers targeting neoplastic lesions in the brain has been investigated in several studies. Based on a literature research regarding experimental FET studies and corresponding clinical applications this article summarizes the knowledge on the uptake behavior of FET, which has been collected in more than 30 experimental studies during the last two decades and discusses the role of these results in the clinical context.
Collapse
Affiliation(s)
- Carina Stegmayr
- Institute of Neuroscience and Medicine 4, Forschungszentrum Juelich, Juelich, Germany
| | - Antje Willuweit
- Institute of Neuroscience and Medicine 4, Forschungszentrum Juelich, Juelich, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine 4, Forschungszentrum Juelich, Juelich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine 4, Forschungszentrum Juelich, Juelich, Germany.,Department of Nuclear Medicine, University of Aachen, Aachen, Germany.,Juelich-Aachen Research Alliance (JARA) - Section JARA-Brain, Juelich, Germany
| |
Collapse
|
16
|
Liesche F, Lukas M, Preibisch C, Shi K, Schlegel J, Meyer B, Schwaiger M, Zimmer C, Förster S, Gempt J, Pyka T. 18F-Fluoroethyl-tyrosine uptake is correlated with amino acid transport and neovascularization in treatment-naive glioblastomas. Eur J Nucl Med Mol Imaging 2019; 46:2163-2168. [PMID: 31289907 DOI: 10.1007/s00259-019-04407-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/18/2019] [Indexed: 01/11/2023]
Abstract
PURPOSE To investigate the in vivo correlation between 18F-fluoroethyl-tyrosine (18F-FET) uptake and amino acid transporter expression and vascularization in treatment-naive glioblastomas. METHODS A total of 43 stereotactic biopsies were obtained from 13 patients with suspected glioblastoma prior to therapy. All patients underwent a dynamic 18F-FET PET/MRI scan before biopsy. Immunohistochemistry was performed using antibodies against SLC7A5 (amino acid transporter), MIB-1 (Ki67, proliferation), CD31 (vascularization) and CA-IX (hypoxia). The intensity of staining was correlated with 18F-FET uptake and the dynamic 18F-FET uptake slope at the biopsy target point. RESULTS In all patients, the final diagnosis was IDH-wildtype glioblastoma, WHO grade IV. Static 18F-FET uptake was significantly correlated with SLC7A5 staining (r = 0.494, p = 0.001). While the dynamic 18F-FET uptake slope did not show a significant correlation with amino acid transporter expression, it was significantly correlated with the number of CD31-positive vessels (r = -0.350, p = 0.031), which is line with earlier results linking 18F-FET kinetics with vascularization and perfusion. Besides, static 18F-FET uptake also showed correlations with CA-IX staining (r = 0.394, p = 0.009) and CD31 positivity (r = 0.410, p = 0.006). While the correlation between static 18F-FET uptake and SLC7A5 staining was confirmed as significant in multivariate analysis, this was not the case for the correlation with CD31 positivity, most likely because of the lower effect size and the relatively low number of samples. No significant correlation between 18F-FET uptake and Ki67 proliferation index was observed in our cohort. CONCLUSION Our results support the findings of preclinical studies suggesting that specific 18F-FET uptake in glioblastomas is mediated by amino acid transporters. As proposed previously, dynamic 18F-FET parameters might be more influenced by perfusion and therefore related to properties of the tumour neovascularization.
Collapse
Affiliation(s)
- Friederike Liesche
- Department of Neuropathology, Institute of Pathology, Technische Universität München, Trogerstraße 18, 81675, Munich, Germany
| | - Mathias Lukas
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany.,Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,Siemens Healthcare GmbH, Berlin, Germany
| | - Christine Preibisch
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Kuangyu Shi
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany.,Department of Nuclear Medicine, Universität Bern, Hochschulstraße 6, 3012, Bern, Switzerland
| | - Jürgen Schlegel
- Department of Neuropathology, Institute of Pathology, Technische Universität München, Trogerstraße 18, 81675, Munich, Germany
| | - Bernhard Meyer
- Department of Neurosurgery, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Claus Zimmer
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Stefan Förster
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Jens Gempt
- Department of Neurosurgery, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Thomas Pyka
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany. .,Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany.
| |
Collapse
|
17
|
Influence of Dexamethasone on O-(2-[ 18F]-Fluoroethyl)-L-Tyrosine Uptake in the Human Brain and Quantification of Tumor Uptake. Mol Imaging Biol 2019; 21:168-174. [PMID: 29845426 DOI: 10.1007/s11307-018-1221-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
PURPOSE O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) is an established positron emission tomography (PET) tracer for brain tumor imaging. This study explores the influence of dexamethasone therapy on [18F]FET uptake in the normal brain and its influence on the maximum and mean tumor-to-brain ratio (TBR). PROCEDURES [18F]FET PET scans of 160 brain tumor patients were evaluated (80 dexamethasone treated, 80 untreated; each group with 40 men/40 women). The standardized uptake value of [18F]FET uptake in the normal brain (SUVbrain) in the different groups was compared. Nine patients were examined repeatedly with and without dexamethasone therapy. RESULTS SUVbrain of [18F]FET uptake was significantly higher in dexamethasone-treated patients than in untreated patients (SUVbrain 1.33 ± 0.1 versus 1.06 ± 0.16 in male and 1.45 ± 0.25 versus 1.31 ± 0.28 in female patients). Similar results were observed in patients with serial PET scans. Furthermore, compared to men, a significantly higher SUVbrain was found in women, both with and without dexamethasone treatment. There were no significant differences between the different groups for TBRmax and TBRmean, which could have been masked by the high standard deviation. In a patient with a stable brain metastasis investigated twice with and without dexamethasone, the TBRmax and the biological tumor volume (BTV) decreased considerably after dexamethasone due to an increased SUVbrain. CONCLUSION Dexamethasone treatment appears to increase the [18F]FET uptake in the normal brain. An effect on TBRmax, TBRmean, and BTV cannot be excluded which should be considered especially for treatment monitoring and the estimation of BTV using [18F]FET PET.
Collapse
|
18
|
Verburg N, Koopman T, Yaqub M, Hoekstra OS, Lammertsma AA, Schwarte LA, Barkhof F, Pouwels PJW, Heimans JJ, Reijneveld JC, Rozemuller AJM, Vandertop WP, Wesseling P, Boellaard R, de Witt Hamer PC. Direct comparison of [ 11C] choline and [ 18F] FET PET to detect glioma infiltration: a diagnostic accuracy study in eight patients. EJNMMI Res 2019; 9:57. [PMID: 31254208 PMCID: PMC6598977 DOI: 10.1186/s13550-019-0523-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/28/2019] [Indexed: 02/07/2023] Open
Abstract
Background Positron emission tomography (PET) is increasingly used to guide local treatment in glioma. The purpose of this study was a direct comparison of two potential tracers for detecting glioma infiltration, O-(2-[18F]-fluoroethyl)-l-tyrosine ([18F] FET) and [11C] choline. Methods Eight consecutive patients with newly diagnosed diffuse glioma underwent dynamic [11C] choline and [18F] FET PET scans. Preceding craniotomy, multiple stereotactic biopsies were obtained from regions inside and outside PET abnormalities. Biopsies were assessed independently for tumour presence by two neuropathologists. Imaging measurements were derived at the biopsy locations from 10 to 40 min [11C] choline and 20–40, 40–60 and 60–90 min [18F] FET intervals, as standardized uptake value (SUV) and tumour-to-brain ratio (TBR). Diagnostic accuracies of both tracers were compared using receiver operating characteristic analysis and generalized linear mixed modelling with consensus histopathological assessment as reference. Results Of the 74 biopsies, 54 (73%) contained tumour. [11C] choline SUV and [18F] FET SUV and TBR at all intervals were higher in tumour than in normal samples. For [18F] FET, the diagnostic accuracy of TBR was higher than that of SUV for intervals 40–60 min (area under the curve: 0.88 versus 0.81, p = 0.026) and 60–90 min (0.90 versus 0.81, p = 0.047). The diagnostic accuracy of [18F] FET TBR 60–90 min was higher than that of [11C] choline SUV 20–40 min (0.87 versus 0.67, p = 0.005). Conclusions [18F] FET was more accurate than [11C] choline for detecting glioma infiltration. Highest accuracy was found for [18F] FET TBR for the interval 60–90 min post-injection. Electronic supplementary material The online version of this article (10.1186/s13550-019-0523-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Niels Verburg
- Neurosurgical Center Amsterdam, Brain Tumour Center Amsterdam, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Thomas Koopman
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Maqsood Yaqub
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Otto S Hoekstra
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Lothar A Schwarte
- Department of Anaesthesiology, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Frederik Barkhof
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.,UCL institutes of Neurology & Healthcare Engineering, Gower St, Bloomsbury, London, WC1E 6BT, UK
| | - Petra J W Pouwels
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Jan J Heimans
- Department of Neurology, Brain Tumour Center Amsterdam, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Jaap C Reijneveld
- Department of Neurology, Brain Tumour Center Amsterdam, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Annemieke J M Rozemuller
- Department of Pathology, Brain Tumour Center Amsterdam, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - William P Vandertop
- Neurosurgical Center Amsterdam, Brain Tumour Center Amsterdam, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, Brain Tumour Center Amsterdam, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.,Princess Máxima Center for Paediatric Oncology, and Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Ronald Boellaard
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Philip C de Witt Hamer
- Neurosurgical Center Amsterdam, Brain Tumour Center Amsterdam, Amsterdam UMC, location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
19
|
Sah BR, Sommerauer M, Mu L, Gonzalez GP, Geistlich S, Treyer V, Schibli R, Buck A, Warnock G, Ametamey SM. Radiation dosimetry of [ 18F]-PSS232-a PET radioligand for imaging mGlu5 receptors in humans. EJNMMI Res 2019; 9:56. [PMID: 31240594 PMCID: PMC6593000 DOI: 10.1186/s13550-019-0522-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 05/28/2019] [Indexed: 11/25/2022] Open
Abstract
Purpose (E)-3-(pyridin-2-ylethynyl)cyclohex-2-enone O-(3-(2-[18F]-fluoroethoxy)propyl) oxime ([18F]-PSS232) is a new PET tracer for imaging of metabotropic glutamate receptor subtype 5 (mGlu5), and has shown promising results in rodents and humans. The aim of this study was to estimate the radiation dosimetry and biodistribution in humans, to assess dose-limiting organs, and to demonstrate safety and tolerability of [18F]-PSS232 in healthy volunteers. Methods PET/CT scans of six healthy male volunteers (mean age 23.5 ± 1.7; 21–26 years) were obtained after intravenous administration of 243 ± 3 MBq of [18F]-PSS232. Serial whole-body (vertex to mid-thigh) PET scans were assessed at ten time points, up to 90 min after tracer injection. Calculation of tracer kinetics and cumulated organ activities were performed using PMOD 3.7 software. Dosimetry estimates were calculated using the OLINDA/EXM software. Results Injection of [18F]-PSS232 was safe and well tolerated. Organs with highest absorbed doses were the gallbladder wall (0.2295 mGy/MBq), liver (0.0547 mGy/MBq), and the small intestine (0.0643 mGy/MBq). Mean effective dose was 3.72 ± 0.12 mSv/volunteer (range 3.61–3.96 mSv; 0.0153 mSv/MBq). Conclusion [18F]-PSS232, a novel [18F]-labeled mGlu5 tracer, showed favorable dosimetry values. Additionally, the tracer was safe and well tolerated.
Collapse
Affiliation(s)
- Bert-Ram Sah
- Department of Nuclear Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland.,Department of Diagnostic, Interventional, and Pediatric Radiology, Inselspital, University of Bern, Bern, Switzerland
| | - Michael Sommerauer
- Department of Nuclear Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland.,Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Linjing Mu
- Department of Nuclear Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Gloria Pla Gonzalez
- Radiopharmaceutical Science, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Susanne Geistlich
- Radiopharmaceutical Science, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Valerie Treyer
- Department of Nuclear Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Roger Schibli
- Radiopharmaceutical Science, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Alfred Buck
- Department of Nuclear Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Geoffrey Warnock
- Department of Nuclear Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland. .,PMOD Technologies LLC, Zurich, Switzerland.
| | - Simon M Ametamey
- Radiopharmaceutical Science, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
20
|
Verhoeven J, Hulpia F, Kersemans K, Bolcaen J, De Lombaerde S, Goeman J, Descamps B, Hallaert G, Van den Broecke C, Deblaere K, Vanhove C, Van der Eycken J, Van Calenbergh S, Goethals I, De Vos F. New fluoroethyl phenylalanine analogues as potential LAT1-targeting PET tracers for glioblastoma. Sci Rep 2019; 9:2878. [PMID: 30814660 PMCID: PMC6393465 DOI: 10.1038/s41598-019-40013-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 02/07/2019] [Indexed: 02/07/2023] Open
Abstract
The use of O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) as a positron emission tomography (PET) tracer for brain tumor imaging might have some limitations because of the relatively low affinity for the L-type amino acid transporter 1 (LAT1). To assess the stereospecificity and evaluate the influence of aromatic ring modification of phenylalanine LAT1 targeting tracers, six different fluoroalkylated phenylalanine analogues were synthesized. After in vitro Ki determination, the most promising compound, 2-[18F]-2-fluoroethyl-L-phenylalanine (2-[18F]FELP), was selected for further evaluation and in vitro comparison with [18F]FET. Subsequently, 2-[18F]FELP was assessed in vivo and compared with [18F]FET and [18F]FDG in a F98 glioblastoma rat model. 2-[18F]FELP showed improved in vitro characteristics over [18F]FET, especially when the affinity and specificity for system L is concerned. Based on our results, 2-[18F]FELP is a promising new PET tracer for brain tumor imaging.
Collapse
Affiliation(s)
| | - Fabian Hulpia
- Laboratory for Medicinal Chemistry, Ghent University, Ghent, Belgium
| | - Ken Kersemans
- Ghent University Hospital, Department of Nuclear Medicine, Ghent, Belgium
| | - Julie Bolcaen
- Ghent University Hospital, Department of Nuclear Medicine, Ghent, Belgium
| | | | - Jan Goeman
- Laboratory for Organic and Bio-organic synthesis, Ghent University, Ghent, Belgium
| | - Benedicte Descamps
- IBiTech-MEDISIP Ghent University, Department of Electronics and Information Systems, Ghent, Belgium
| | - Giorgio Hallaert
- Ghent University Hospital, Department of Neurosurgery, Ghent, Belgium
| | | | - Karel Deblaere
- Ghent University Hospital, Department of Radiology and Medical Imaging, Ghent, Belgium
| | - Christian Vanhove
- IBiTech-MEDISIP Ghent University, Department of Electronics and Information Systems, Ghent, Belgium
| | - Johan Van der Eycken
- Laboratory for Organic and Bio-organic synthesis, Ghent University, Ghent, Belgium
| | | | - Ingeborg Goethals
- Ghent University Hospital, Department of Nuclear Medicine, Ghent, Belgium
| | - Filip De Vos
- Laboratory of Radiopharmacy, Ghent University, Ghent, Belgium
| |
Collapse
|
21
|
Peyraga G, Robaine N, Khalifa J, Cohen-Jonathan-Moyal E, Payoux P, Laprie A. Molecular PET imaging in adaptive radiotherapy: brain. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2018; 62:337-348. [PMID: 30497232 DOI: 10.23736/s1824-4785.18.03116-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Owing to their heterogeneity and radioresistance, the prognosis of primitive brain tumors, which are mainly glial tumors, remains poor. Dose escalation in radioresistant areas is a potential issue for improving local control and overall survival. This review focuses on advances in biological and metabolic imaging of brain tumors that are proving to be essential for defining tumor target volumes in radiation therapy (RT) and for increasing the use of DPRT (dose painting RT) and ART (adaptative RT), to optimize dose in radio-resistant areas. EVIDENCE ACQUISITION Various biological imaging modalities such as PET (hypoxia, glucidic metabolism, protidic metabolism, cellular proliferation, inflammation, cellular membrane synthesis) and MRI (spectroscopy) may be used to identify these areas of radioresistance. The integration of these biological imaging modalities improves the diagnosis, prognosis and treatment of brain tumors. EVIDENCE SYNTHESIS Technological improvements (PET and MRI), the development of research, and intensive cooperation between different departments are necessary before using daily metabolic imaging (PET and MRI) to treat patients with brain tumors. CONCLUSIONS The adaptation of treatment volumes during RT (ART) seems promising, but its development requires improvements in several areas and an interdisciplinary approach involving radiology, nuclear medicine and radiotherapy. We review the literature on biological imaging to outline the perspectives for using DPRT and ART in brain tumors.
Collapse
Affiliation(s)
- Guillaume Peyraga
- Department of Radiation Therapy, Claudius Regaud Institute, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Nesrine Robaine
- Department of Nuclear Medicine, Claudius Regaud Institute, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Jonathan Khalifa
- Department of Radiation Therapy, Claudius Regaud Institute, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France.,Paul Sabatier University, Toulouse III, Toulouse, France
| | - Elizabeth Cohen-Jonathan-Moyal
- Department of Radiation Therapy, Claudius Regaud Institute, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France.,Paul Sabatier University, Toulouse III, Toulouse, France
| | - Pierre Payoux
- Department of Nuclear Medicine, Purpan University Hospital Center, Toulouse, France
| | - Anne Laprie
- Department of Radiation Therapy, Claudius Regaud Institute, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France - .,Paul Sabatier University, Toulouse III, Toulouse, France
| |
Collapse
|
22
|
Koopman T, Verburg N, Schuit RC, Pouwels PJW, Wesseling P, Windhorst AD, Hoekstra OS, de Witt Hamer PC, Lammertsma AA, Boellaard R, Yaqub M. Quantification of O-(2-[ 18F]fluoroethyl)-L-tyrosine kinetics in glioma. EJNMMI Res 2018; 8:72. [PMID: 30066053 PMCID: PMC6068050 DOI: 10.1186/s13550-018-0418-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/27/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND This study identified the optimal tracer kinetic model for quantification of dynamic O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) positron emission tomography (PET) studies in seven patients with diffuse glioma (four glioblastoma, three lower grade glioma). The performance of more simplified approaches was evaluated by comparison with the optimal compartment model. Additionally, the relationship with cerebral blood flow-determined by [15O]H2O PET-was investigated. RESULTS The optimal tracer kinetic model was the reversible two-tissue compartment model. Agreement analysis of binding potential estimates derived from reference tissue input models with the distribution volume ratio (DVR)-1 derived from the plasma input model showed no significant average difference and limits of agreement of - 0.39 and 0.37. Given the range of DVR-1 (- 0.25 to 1.5), these limits are wide. For the simplified methods, the 60-90 min tumour-to-blood ratio to parent plasma concentration yielded the highest correlation with volume of distribution VT as calculated by the plasma input model (r = 0.97). The 60-90 min standardized uptake value (SUV) showed better correlation with VT (r = 0.77) than SUV based on earlier intervals. The 60-90 min SUV ratio to contralateral healthy brain tissue showed moderate agreement with DVR with no significant average difference and limits of agreement of - 0.24 and 0.30. A significant but low correlation was found between VT and CBF in the tumour regions (r = 0.61, p = 0.007). CONCLUSION Uptake of [18F]FET was best modelled by a reversible two-tissue compartment model. Reference tissue input models yielded estimates of binding potential which did not correspond well with plasma input-derived DVR-1. In comparison, SUV ratio to contralateral healthy brain tissue showed slightly better performance, if measured at the 60-90 min interval. SUV showed only moderate correlation with VT. VT shows correlation with CBF in tumour.
Collapse
Affiliation(s)
- Thomas Koopman
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Niels Verburg
- Neurosurgical Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
- Brain Tumor Center Amsterdam, Amsterdam, The Netherlands
| | - Robert C. Schuit
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Petra J. W. Pouwels
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Pathology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Albert D. Windhorst
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Otto S. Hoekstra
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Philip C. de Witt Hamer
- Neurosurgical Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
- Brain Tumor Center Amsterdam, Amsterdam, The Netherlands
| | - Adriaan A. Lammertsma
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Groningen, The Netherlands
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| |
Collapse
|
23
|
Parent EE, Schuster DM. Update on 18F-Fluciclovine PET for Prostate Cancer Imaging. J Nucl Med 2018; 59:733-739. [PMID: 29523631 PMCID: PMC6910635 DOI: 10.2967/jnumed.117.204032] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/08/2018] [Indexed: 01/22/2023] Open
Abstract
PET is a functional imaging method that can exploit various aspects of tumor biology to enable greater detection of prostate cancer than can be provided by morphologic imaging alone. Anti-1-amino-3-18F-flurocyclobutane-1-carboxylic acid (18F-fluciclovine) is a nonnaturally occurring amino acid PET radiotracer that was recently approved by the U.S. Food and Drug Administration for detection of suspected recurrent prostate cancer. The tumor-imaging features of this radiotracer mirror the upregulation of transmembrane amino acid transport that occurs in prostate cancer because of increased amino acid metabolism for energy and protein synthesis. This continuing medical education article provides an overview on 18F-fluciclovine PET diagnostic capabilities for primary and metastatic disease, including reviews of published comparisons to conventional imaging and other molecular imaging agents. Additionally, the imaging procedure and image interpretation are detailed, including physiologic and pathologic uptake patterns and pitfalls.
Collapse
Affiliation(s)
- Ephraim E Parent
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - David M Schuster
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
24
|
Schröder S, Wenzel B, Deuther-Conrad W, Teodoro R, Kranz M, Scheunemann M, Egerland U, Höfgen N, Briel D, Steinbach J, Brust P. Investigation of an 18F-labelled Imidazopyridotriazine for Molecular Imaging of Cyclic Nucleotide Phosphodiesterase 2A. Molecules 2018; 23:molecules23030556. [PMID: 29498659 PMCID: PMC6017663 DOI: 10.3390/molecules23030556] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/15/2018] [Accepted: 02/23/2018] [Indexed: 11/16/2022] Open
Abstract
Specific radioligands for in vivo visualization and quantification of cyclic nucleotide phosphodiesterase 2A (PDE2A) by positron emission tomography (PET) are increasingly gaining interest in brain research. Herein we describe the synthesis, the 18F-labelling as well as the biological evaluation of our latest PDE2A (radio-)ligand 9-(5-Butoxy-2-fluorophenyl)-2-(2-([18F])fluoroethoxy)-7-methylimidazo[5,1-c]pyrido[2,3-e][1,2,4]triazine (([18F])TA5). It is the most potent PDE2A ligand out of our series of imidazopyridotriazine-based derivatives so far (IC50 hPDE2A = 3.0 nM; IC50 hPDE10A > 1000 nM). Radiolabelling was performed in a one-step procedure starting from the corresponding tosylate precursor. In vitro autoradiography on rat and pig brain slices displayed a homogenous and non-specific binding of the radioligand. Investigation of stability in vivo by reversed-phase HPLC (RP-HPLC) and micellar liquid chromatography (MLC) analyses of plasma and brain samples obtained from mice revealed a high fraction of one main radiometabolite. Hence, we concluded that [18F]TA5 is not appropriate for molecular imaging of PDE2A neither in vitro nor in vivo. Our ongoing work is focusing on further structurally modified compounds with enhanced metabolic stability.
Collapse
Affiliation(s)
- Susann Schröder
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Leipzig 04318, Germany; (B.W.); (W.D.-C.); (R.T.); (M.K.); (M.S.); (J.S.); (P.B.)
- Correspondence: ; Tel.: +49-341-234-179-4631
| | - Barbara Wenzel
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Leipzig 04318, Germany; (B.W.); (W.D.-C.); (R.T.); (M.K.); (M.S.); (J.S.); (P.B.)
| | - Winnie Deuther-Conrad
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Leipzig 04318, Germany; (B.W.); (W.D.-C.); (R.T.); (M.K.); (M.S.); (J.S.); (P.B.)
| | - Rodrigo Teodoro
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Leipzig 04318, Germany; (B.W.); (W.D.-C.); (R.T.); (M.K.); (M.S.); (J.S.); (P.B.)
| | - Mathias Kranz
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Leipzig 04318, Germany; (B.W.); (W.D.-C.); (R.T.); (M.K.); (M.S.); (J.S.); (P.B.)
| | - Matthias Scheunemann
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Leipzig 04318, Germany; (B.W.); (W.D.-C.); (R.T.); (M.K.); (M.S.); (J.S.); (P.B.)
| | - Ute Egerland
- BioCrea GmbH, Radebeul 01445, Germany; (U.E.); (N.H.)
| | | | - Detlef Briel
- Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Faculty of Medicine, Leipzig University, Leipzig 04103, Germany;
| | - Jörg Steinbach
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Leipzig 04318, Germany; (B.W.); (W.D.-C.); (R.T.); (M.K.); (M.S.); (J.S.); (P.B.)
| | - Peter Brust
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Leipzig 04318, Germany; (B.W.); (W.D.-C.); (R.T.); (M.K.); (M.S.); (J.S.); (P.B.)
| |
Collapse
|
25
|
Evaluation of factors influencing 18F-FET uptake in the brain. NEUROIMAGE-CLINICAL 2017; 17:491-497. [PMID: 29159062 PMCID: PMC5684535 DOI: 10.1016/j.nicl.2017.11.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 10/24/2017] [Accepted: 11/07/2017] [Indexed: 01/20/2023]
Abstract
PET using the amino-acid O-(2-18F-fluoroethyl)-l-tyrosine (18F-FET) is gaining increasing interest for brain tumour management. Semi-quantitative analysis of tracer uptake in brain tumours is based on the standardized uptake value (SUV) and the tumour-to-brain ratio (TBR). The aim of this study was to explore physiological factors that might influence the relationship of SUV of 18F-FET uptake in various brain areas, and thus affect quantification of 18F-FET uptake in brain tumours. Negative 18F-FET PET scans of 107 subjects, showing an inconspicuous brain distribution of 18F-FET, were evaluated retrospectively. Whole-brain quantitative analysis with Statistical Parametric Mapping (SPM) using parametric SUV PET images, and volumes of interest (VOIs) analysis with fronto-parietal, temporal, occipital, and cerebellar SUV background areas were performed to study the effect of age, gender, height, weight, injected activity, body mass index (BMI), and body surface area (BSA). After multivariate analysis, female gender and high BMI were found to be two independent factors associated with increased SUV of 18F-FET uptake in the brain. In women, SUVmean of 18F-FET uptake in the brain was 23% higher than in men (p < 0.01). SUVmean of 18F-FET uptake in the brain was positively correlated with BMI (r = 0.29; p < 0.01). The influence of these factors on SUV of 18F-FET was similar in all brain areas. In conclusion, SUV of 18F-FET in the normal brain is influenced by gender and weakly by BMI, but changes are similar in all brain areas. SUVmean of 18F-FET in the normal brain is influenced by gender. SUVmean of 18F-FET in the normal brain is weekly influenced by BMI. The influence of these factors on SUV of 18F-FET is similar in all brain areas.
Collapse
|
26
|
Imaging of amino acid transport in brain tumours: Positron emission tomography with O-(2-[ 18 F]fluoroethyl)- L -tyrosine (FET). Methods 2017; 130:124-134. [DOI: 10.1016/j.ymeth.2017.05.019] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/08/2017] [Accepted: 05/21/2017] [Indexed: 01/01/2023] Open
|
27
|
Imperiale A, Boisson F, Kreutter G, Goichot B, Namer IJ, Bachellier P, Laquerriere P, Kessler L, Marchand P, Brasse D. O-(2- 18F-fluoroethyl)-l-tyrosine ( 18F-FET) uptake in insulinoma: first results from a xenograft mouse model and from human. Nucl Med Biol 2017; 53:21-28. [PMID: 28793277 DOI: 10.1016/j.nucmedbio.2017.07.005] [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: 06/14/2017] [Revised: 07/05/2017] [Accepted: 07/07/2017] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Herein we have evaluated the uptake of O-(2-18F-fluoroethyl)-l-tyrosine (18F-FET) in insulinoma in comparison with those of 6-18F-fluoro-3,4-dihydroxy-l-phenylalanine (18F-FDOPA) providing first data from both murine xenograft model and one patient with proved endogenous hyperinsulinemic hypoglycemia. METHODS Dynamic 18F-FET and carbidopa-assisted 18F-FDOPA PET were performed on tumor-bearing nude mice after subcutaneous injection of RIN-m5F murine beta cells and on a 30-year-old man with type-1 multiple endocrine neoplasia and hyperinsulinemic hypoglycemia defined by a positive fasting test. RESULTS Seven and three nude mice bearing a RIN-m5F insulinoma xenograft were respectively studied by 18F-FET and 18F-FDOPA μPET. Insulinoma xenograft was detected in all the imaged animals. Xenograft was characterized by an early but moderate increase of 18F-FET uptake followed by a slight decline of uptake intensity during the 20 min dynamic acquisition. Tumoral radiotracer peak intensity and the highest tumor-to-background contrast were reached about 5 minutes after 18F-FET iv. injection (mean SUV: 1.21 ± 0.10). The biodistribution of 18F-FET and 18F-FDOPA and their dynamic tumoral uptake profile and intensity were similar. In the examined patient, 18F-FDOPA and 18F-FET PET/CT showed one concordant focal area of well-defined increased uptake in the pancreatic tail corresponding to 11 mm histologically proved insulinoma. The SUVmax tumor to liver ratio was 1.5, 1.1 for 18F-FDOPA, 1.1, 1 for 18F-FET at early (0-5 min post injection) and delayed (5-20 min post injection) PET/CT acquisition, respectively. Despite the relatively low tumoral uptake intensity, insulinoma was clearly identified due to the low background in the pancreas. At the contrary, no 18F-FDOPA or 18F-FET tumoral uptake was revealed on whole-body PET/CT images performed about 30 min after radiotracer administration. Note of worth, the dynamic uptake pattern of 18F-FET and 18F-FDOPA were similar between human insulinoma and mice xenograft tumor. CONCLUSION 18F-FET PET compared equally to 18F-FDOPA PET in a preclinical RIN-m5F murine model of insulinoma and in one patient with insulinoma-related hypoglycemia. However, in both cases, the tumoral uptake intensity was moderate and the tumor was only visible until 20 min after radiotracer injection. Hence, caution should be taken before asserting the translational relevance of our results in the clinical practices. However, the structural analogies between 18F-FET and 18F-FDOPA as well as the limited pancreatic uptake of 18F-FET in human, encourage evaluating 18F-FET as diagnostic radiotracer for insulinoma detection in further prospective studies involving large cohorts of patients.
Collapse
Affiliation(s)
- Alessio Imperiale
- Biophysics and Nuclear Medicine, Strasbourg University Hospitals, Strasbourg, France; ICube, CNRS/UMR 7357, Strasbourg University, Strasbourg, France; Federation of Translational Medicine of Strasbourg (FMTS), Faculty of Medicine, Strasbourg University, Strasbourg, France; Université de Strasbourg, CNRS, IPHC, UMR 7178, F-67000, Strasbourg, France.
| | - Frédéric Boisson
- Université de Strasbourg, CNRS, IPHC, UMR 7178, F-67000, Strasbourg, France
| | - Guillaume Kreutter
- Federation of Translational Medicine of Strasbourg (FMTS), Faculty of Medicine, Strasbourg University, Strasbourg, France; EA7293, Vascular and Tissular Stress in Transplantation, Illkirch, France
| | - Bernard Goichot
- Internal Medicine, Strasbourg University Hospitals, Strasbourg, France
| | - Izzie Jacques Namer
- Biophysics and Nuclear Medicine, Strasbourg University Hospitals, Strasbourg, France; ICube, CNRS/UMR 7357, Strasbourg University, Strasbourg, France; Federation of Translational Medicine of Strasbourg (FMTS), Faculty of Medicine, Strasbourg University, Strasbourg, France
| | - Philippe Bachellier
- Visceral Surgery and Transplantation, Strasbourg University Hospitals, Strasbourg, France
| | | | - Laurence Kessler
- Federation of Translational Medicine of Strasbourg (FMTS), Faculty of Medicine, Strasbourg University, Strasbourg, France; EA7293, Vascular and Tissular Stress in Transplantation, Illkirch, France; Diabetology, Strasbourg University Hospitals, Strasbourg, France
| | - Patrice Marchand
- Université de Strasbourg, CNRS, IPHC, UMR 7178, F-67000, Strasbourg, France
| | - David Brasse
- Université de Strasbourg, CNRS, IPHC, UMR 7178, F-67000, Strasbourg, France
| |
Collapse
|
28
|
Assessment of Amino Acid/Drug Transporters for Renal Transport of [ 18F]Fluciclovine (anti-[ 18F]FACBC) in Vitro. Int J Mol Sci 2016; 17:ijms17101730. [PMID: 27754421 PMCID: PMC5085761 DOI: 10.3390/ijms17101730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/14/2016] [Accepted: 10/08/2016] [Indexed: 01/19/2023] Open
Abstract
[18F]Fluciclovine (trans-1-amino-3-[18F]fluorocyclobutanecarboxylic acid; anti-[18F]FACBC), a positron emission tomography tracer used for the diagnosis of recurrent prostate cancer, is transported via amino acid transporters (AATs) with high affinity (Km: 97-230 μM). However, the mechanism underlying urinary excretion is unknown. In this study, we investigated the involvement of AATs and drug transporters in renal [18F]fluciclovine reuptake. [14C]Fluciclovine (trans-1-amino-3-fluoro[1-14C]cyclobutanecarboxylic acid) was used because of its long half-life. The involvement of AATs in [14C]fluciclovine transport was measured by apical-to-basal transport using an LLC-PK1 monolayer as model for renal proximal tubules. The contribution of drug transporters herein was assessed using vesicles/cells expressing the drug transporters P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), multidrug resistance-associated protein 4 (MRP4), organic anion transporter 1 (OAT1), organic anion transporter 3 (OAT3) , organic cation transporter 2 (OCT2), organic anion transporting polypeptide 1B1 (OATP1B1), and organic anion transporting polypeptide 1B3 (OATP1B3). The apical-to-basal transport of [14C]fluciclovine was attenuated by l-threonine, the substrate for system alanine-serine-cysteine (ASC) AATs. [14C]Fluciclovine uptake by drug transporter-expressing vesicles/cells was not significantly different from that of control vesicles/cells. Fluciclovine inhibited P-gp, MRP4, OAT1, OCT2, and OATP1B1 (IC50 > 2.95 mM). Therefore, system ASC AATs may be partly involved in the renal reuptake of [18F]fluciclovine. Further, given that [18F]fluciclovine is recognized as an inhibitor with millimolar affinity for the tested drug transporters, slow urinary excretion of [18F]fluciclovine may be mediated by system ASC AATs, but not by drug transporters.
Collapse
|
29
|
Kranz M, Sattler B, Wüst N, Deuther-Conrad W, Patt M, Meyer PM, Fischer S, Donat CK, Wünsch B, Hesse S, Steinbach J, Brust P, Sabri O. Evaluation of the Enantiomer Specific Biokinetics and Radiation Doses of [(18)F]Fluspidine-A New Tracer in Clinical Translation for Imaging of σ₁ Receptors. Molecules 2016; 21:E1164. [PMID: 27598110 PMCID: PMC6273209 DOI: 10.3390/molecules21091164] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 08/22/2016] [Accepted: 08/26/2016] [Indexed: 11/17/2022] Open
Abstract
The enantiomers of [(18)F]fluspidine, recently developed for imaging of σ₁ receptors, possess distinct pharmacokinetics facilitating their use in different clinical settings. To support their translational potential, we estimated the human radiation dose of (S)-(-)-[(18)F]fluspidine and (R)-(+)-[(18)F]fluspidine from ex vivo biodistribution and PET/MRI data in mice after extrapolation to the human scale. In addition, we validated the preclinical results by performing a first-in-human PET/CT study using (S)-(-)-[(18)F]fluspidine. Based on the respective time-activity curves, we calculated using OLINDA the particular organ doses (ODs) and effective doses (EDs). The ED values of (S)-(-)-[(18)F]fluspidine and (R)-(+)-[(18)F]fluspidine differed significantly with image-derived values obtained in mice with 12.9 μSv/MBq and 14.0 μSv/MBq (p < 0.025), respectively. A comparable ratio was estimated from the biodistribution data. In the human study, the ED of (S)-(-)-[(18)F]fluspidine was calculated as 21.0 μSv/MBq. Altogether, the ED values for both [(18)F]fluspidine enantiomers determined from the preclinical studies are comparable with other (18)F-labeled PET imaging agents. In addition, the first-in-human study confirmed that the radiation risk of (S)-(-)-[(18)F]fluspidine imaging is within acceptable limits. However, as already shown for other PET tracers, the actual ED of (S)-(-)-[(18)F]fluspidine in humans was underestimated by preclinical imaging which needs to be considered in other first-in-human studies.
Collapse
Affiliation(s)
- Mathias Kranz
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Leipzig 04318, Germany.
| | - Bernhard Sattler
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig 04103, Germany.
| | - Nathanael Wüst
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig 04103, Germany.
| | - Winnie Deuther-Conrad
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Leipzig 04318, Germany.
| | - Marianne Patt
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig 04103, Germany.
| | - Philipp M Meyer
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig 04103, Germany.
| | - Steffen Fischer
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Leipzig 04318, Germany.
| | - Cornelius K Donat
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Leipzig 04318, Germany.
- Division of Brain Sciences, Department of Medicine, Hammersmith Hospital Campus, Imperial College London, London SW7 2AZ, UK.
| | - Bernhard Wünsch
- Pharmaceutical and Medicinal Chemistry, University Münster, Münster 48149, Germany.
| | - Swen Hesse
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig 04103, Germany.
- Integrated Research and Treatment Center (IFB) Adiposity Diseases, University Hospital Leipzig, Leipzig 04103, Germany.
| | - Jörg Steinbach
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Leipzig 04318, Germany.
| | - Peter Brust
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Leipzig 04318, Germany.
| | - Osama Sabri
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig 04103, Germany.
| |
Collapse
|
30
|
Hachem M, Tiberi M, Ismail B, Hunter CR, Arksey N, Hadizad T, Beanlands RS, deKemp RA, DaSilva JN. Characterization of 18F-FPyKYNE-Losartan for Imaging AT1 Receptors. J Nucl Med 2016; 57:1612-1617. [DOI: 10.2967/jnumed.115.170951] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 04/11/2016] [Indexed: 12/25/2022] Open
|
31
|
Liu J, Wenzel B, Dukic-Stefanovic S, Teodoro R, Ludwig FA, Deuther-Conrad W, Schröder S, Chezal JM, Moreau E, Brust P, Maisonial-Besset A. Development of a New Radiofluorinated Quinoline Analog for PET Imaging of Phosphodiesterase 5 (PDE5) in Brain. Pharmaceuticals (Basel) 2016; 9:E22. [PMID: 27110797 PMCID: PMC4932540 DOI: 10.3390/ph9020022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/12/2016] [Accepted: 04/13/2016] [Indexed: 11/16/2022] Open
Abstract
Phosphodiesterases (PDEs) are enzymes that play a major role in cell signalling by hydrolysing the secondary messengers cyclic adenosine monophosphate (cAMP) and/or cyclic guanosine monophosphate (cGMP) throughout the body and brain. Altered cyclic nucleotide-mediated signalling has been associated with a wide array of disorders, including neurodegenerative disorders. Recently, PDE5 has been shown to be involved in neurodegenerative disorders such as Alzheimer's disease, but its precise role has not been elucidated yet. To visualize and quantify the expression of this enzyme in brain, we developed a radiotracer for specific PET imaging of PDE5. A quinoline-based lead compound has been structurally modified resulting in the fluoroethoxymethyl derivative ICF24027 with high inhibitory activity towards PDE5 (IC50 = 1.86 nM). Radiolabelling with fluorine-18 was performed by a one-step nucleophilic substitution reaction using a tosylate precursor (RCY(EOB) = 12.9% ± 1.8%; RCP > 99%; SA(EOS) = 70-126 GBq/μmol). In vitro autoradiographic studies of [(18)F]ICF24027 on different mouse tissue as well as on porcine brain slices demonstrated a moderate specific binding to PDE5. In vivo studies in mice revealed that [(18)F]ICF24027 was metabolized under formation of brain penetrable radiometabolites making the radiotracer unsuitable for PET imaging of PDE5 in brain.
Collapse
Affiliation(s)
- Jianrong Liu
- INSERM-Université d'Auvergne, UMR 990, IMTV, BP 184, F-63005 Clermont-Ferrand Cedex, France.
| | - Barbara Wenzel
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Permoserstrasse 15, 04318 Leipzig, Germany.
| | - Sladjana Dukic-Stefanovic
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Permoserstrasse 15, 04318 Leipzig, Germany.
| | - Rodrigo Teodoro
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Permoserstrasse 15, 04318 Leipzig, Germany.
| | - Friedrich-Alexander Ludwig
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Permoserstrasse 15, 04318 Leipzig, Germany.
| | - Winnie Deuther-Conrad
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Permoserstrasse 15, 04318 Leipzig, Germany.
| | - Susann Schröder
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Permoserstrasse 15, 04318 Leipzig, Germany.
| | - Jean-Michel Chezal
- INSERM-Université d'Auvergne, UMR 990, IMTV, BP 184, F-63005 Clermont-Ferrand Cedex, France.
| | - Emmanuel Moreau
- INSERM-Université d'Auvergne, UMR 990, IMTV, BP 184, F-63005 Clermont-Ferrand Cedex, France.
| | - Peter Brust
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Permoserstrasse 15, 04318 Leipzig, Germany.
| | | |
Collapse
|
32
|
Wei L, Tominaga H, Ohgaki R, Wiriyasermkul P, Hagiwara K, Okuda S, Kaira K, Oriuchi N, Nagamori S, Kanai Y. Specific transport of 3-fluoro-l-α-methyl-tyrosine by LAT1 explains its specificity to malignant tumors in imaging. Cancer Sci 2016; 107:347-52. [PMID: 26749017 PMCID: PMC4814262 DOI: 10.1111/cas.12878] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 12/27/2015] [Accepted: 01/02/2016] [Indexed: 12/13/2022] Open
Abstract
3‐18F‐l‐α‐methyl‐tyrosine ([18F]FAMT), a PET probe for tumor imaging, has advantages of high cancer‐specificity and lower physiologic background. FAMT‐PET has been proved useful in clinical studies for the prediction of prognosis, the assessment of therapy response and the differentiation of malignant tumors from inflammation and benign lesions. The tumor uptake of [18F]FAMT in PET is strongly correlated with the expression of L‐type amino acid transporter 1 (LAT1), an isoform of system L upregulated in cancers. In this study, to assess the transporter‐mediated mechanisms in FAMT uptake by tumors, we examined amino acid transporters for FAMT transport. We synthesized [14C]FAMT and measured its transport by human amino acid transporters expressed in Xenopus oocytes. The transport of FAMT was compared with that of l‐methionine, a well‐studied amino acid PET probe. The significance of LAT1 in FAMT uptake by tumor cells was confirmed by siRNA knockdown. Among amino acid transporters, [14C]FAMT was specifically transported by LAT1, whereas l‐[14C]methionine was taken up by most of the transporters. Km of LAT1‐mediated [14C]FAMT transport was 72.7 μM, similar to that for endogenous substrates. Knockdown of LAT1 resulted in the marked reduction of [14C]FAMT transport in HeLa S3 cells, confirming the contribution of LAT1 in FAMT uptake by tumor cells. FAMT is highly specific to cancer‐type amino acid transporter LAT1, which explains the cancer‐specific accumulation of [18F]FAMT in PET. This, vice versa, further supports the cancer‐specific expression of LAT1. This study has established FAMT as a LAT1‐specific molecular probe to monitor the expression of a potential tumor biomarker LAT1.
Collapse
Affiliation(s)
- Ling Wei
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hideyuki Tominaga
- Advanced Clinical Research Center, Fukushima Medical University, Fukushima, Japan
| | - Ryuichi Ohgaki
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Pattama Wiriyasermkul
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Kohei Hagiwara
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Suguru Okuda
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Kyoichi Kaira
- Department of Oncology Clinical Development, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Noboru Oriuchi
- Advanced Clinical Research Center, Fukushima Medical University, Fukushima, Japan
| | - Shushi Nagamori
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
33
|
Wei L, Tominaga H, Ohgaki R, Wiriyasermkul P, Hagiwara K, Okuda S, Kaira K, Kato Y, Oriuchi N, Nagamori S, Kanai Y. Transport of 3-fluoro-l-α-methyl-tyrosine (FAMT) by organic ion transporters explains renal background in [18F]FAMT positron emission tomography. J Pharmacol Sci 2016; 130:101-9. [DOI: 10.1016/j.jphs.2016.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/17/2015] [Accepted: 01/06/2016] [Indexed: 12/21/2022] Open
|
34
|
Dunet V, Pomoni A, Hottinger A, Nicod-Lalonde M, Prior JO. Performance of 18F-FET versus 18F-FDG-PET for the diagnosis and grading of brain tumors: systematic review and meta-analysis. Neuro Oncol 2015; 18:426-34. [PMID: 26243791 DOI: 10.1093/neuonc/nov148] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 07/04/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND For the past decade (18)F-fluoro-ethyl-l-tyrosine (FET) and (18)F-fluoro-deoxy-glucose (FDG) positron emission tomography (PET) have been used for the assessment of patients with brain tumor. However, direct comparison studies reported only limited numbers of patients. Our purpose was to compare the diagnostic performance of FET and FDG-PET. METHODS We examined studies published between January 1995 and January 2015 in the PubMed database. To be included the study should: (i) use FET and FDG-PET for the assessment of patients with isolated brain lesion and (ii) use histology as the gold standard. Analysis was performed on a per patient basis. Study quality was assessed with STARD and QUADAS criteria. RESULTS Five studies (119 patients) were included. For the diagnosis of brain tumor, FET-PET demonstrated a pooled sensitivity of 0.94 (95% CI: 0.79-0.98) and pooled specificity of 0.88 (95% CI: 0.37-0.99), with an area under the curve of 0.96 (95% CI: 0.94-0.97), a positive likelihood ratio (LR+) of 8.1 (95% CI: 0.8-80.6), and a negative likelihood ratio (LR-) of 0.07 (95% CI: 0.02-0.30), while FDG-PET demonstrated a sensitivity of 0.38 (95% CI: 0.27-0.50) and specificity of 0.86 (95% CI: 0.31-0.99), with an area under the curve of 0.40 (95% CI: 0.36-0.44), an LR+ of 2.7 (95% CI: 0.3-27.8), and an LR- of 0.72 (95% CI: 0.47-1.11). Target-to-background ratios of either FDG or FET, however, allow distinction between low- and high-grade gliomas (P > .11). CONCLUSIONS For brain tumor diagnosis, FET-PET performed much better than FDG and should be preferred when assessing a new isolated brain tumor. For glioma grading, however, both tracers showed similar performances.
Collapse
Affiliation(s)
- Vincent Dunet
- Department of Radiology, Lausanne University Hospital, Lausanne, Switzerland (V.D.); Nuclear Medicine, Lausanne University Hospital, Lausanne, Switzerland (A.P., M.N.-L., J.O.P.); Clinical Neurosciences, Lausanne University Hospital, Lausanne, Switzerland (A.H.); Oncology, Lausanne University Hospital, Lausanne, Switzerland (A.H.)
| | - Anastasia Pomoni
- Department of Radiology, Lausanne University Hospital, Lausanne, Switzerland (V.D.); Nuclear Medicine, Lausanne University Hospital, Lausanne, Switzerland (A.P., M.N.-L., J.O.P.); Clinical Neurosciences, Lausanne University Hospital, Lausanne, Switzerland (A.H.); Oncology, Lausanne University Hospital, Lausanne, Switzerland (A.H.)
| | - Andreas Hottinger
- Department of Radiology, Lausanne University Hospital, Lausanne, Switzerland (V.D.); Nuclear Medicine, Lausanne University Hospital, Lausanne, Switzerland (A.P., M.N.-L., J.O.P.); Clinical Neurosciences, Lausanne University Hospital, Lausanne, Switzerland (A.H.); Oncology, Lausanne University Hospital, Lausanne, Switzerland (A.H.)
| | - Marie Nicod-Lalonde
- Department of Radiology, Lausanne University Hospital, Lausanne, Switzerland (V.D.); Nuclear Medicine, Lausanne University Hospital, Lausanne, Switzerland (A.P., M.N.-L., J.O.P.); Clinical Neurosciences, Lausanne University Hospital, Lausanne, Switzerland (A.H.); Oncology, Lausanne University Hospital, Lausanne, Switzerland (A.H.)
| | - John O Prior
- Department of Radiology, Lausanne University Hospital, Lausanne, Switzerland (V.D.); Nuclear Medicine, Lausanne University Hospital, Lausanne, Switzerland (A.P., M.N.-L., J.O.P.); Clinical Neurosciences, Lausanne University Hospital, Lausanne, Switzerland (A.H.); Oncology, Lausanne University Hospital, Lausanne, Switzerland (A.H.)
| |
Collapse
|
35
|
Autio A, Virtanen H, Tolvanen T, Liljenbäck H, Oikonen V, Saanijoki T, Siitonen R, Käkelä M, Schüssele A, Teräs M, Roivainen A. Absorption, distribution and excretion of intravenously injected (68)Ge/ (68)Ga generator eluate in healthy rats, and estimation of human radiation dosimetry. EJNMMI Res 2015; 5:117. [PMID: 26183032 PMCID: PMC4504870 DOI: 10.1186/s13550-015-0117-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 07/06/2015] [Indexed: 11/17/2022] Open
Abstract
Background This study evaluated the absorption, distribution, and excretion of Gallium-68 (68Ga) radionuclide after a single intravenous (i.v.) injection of 68Ge/68Ga generator eluate in healthy rats. Additionally, human radiation doses were estimated from the rat data. Methods Twenty-one female and 21 male Sprague-Dawley rats were i.v. injected with 47 ± 4 MBq of 68Ge/68Ga generator eluate, and the radioactivity of excised organs was measured using a gamma counter at 5, 30, 60, 120, or 180 min afterwards (n = 3–7 for each time point). The radioactivity concentration and plasma pharmacokinetic parameters were calculated. Subsequently, the estimates for human radiation dosimetry were determined. Additionally, 4 female and 5 male rats were positron emission tomography (PET) imaged for in vivo visualization of biodistribution. Results 68Ga radioactivity was cleared relatively slowly from blood circulation and excreted into the urine, with some retention in the liver and spleen. Notably, the 68Ga radioactivity in female genital organs, i.e., the uterus and ovaries, was considerable higher compared with male genitals. Extrapolating from the female and male rat 68Ga data, the estimated effective dose was 0.0308 mSv/MBq for a 57-kg woman and 0.0191 mSv/MBq for a 70-kg man. Conclusions The estimated human radiation burden of the 68Ge/68Ga generator eluate was slightly higher for females and similar for males as compared with somatostatin receptor ligands 68Ga-DOTANOC, 68Ga-DOTATOC, and 68Ga-DOTATATE, which is probably due to the retention in the liver and spleen. Our results revealed some differences between female and male rat data, which, at least in part, may be explained by the small sample size.
Collapse
Affiliation(s)
- Anu Autio
- Turku PET Centre, Turku University Hospital, University of Turku, FI-20521, Turku, Finland,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Tang C, Xu Z, Hu K, Yao B, Tang G, Nie D. Radiation dosimetry estimation of N-(2-[(18)F]fluoropropionyl)- L-glutamate based on the mice distribution data. Appl Radiat Isot 2015; 98:108-112. [PMID: 25661723 DOI: 10.1016/j.apradiso.2015.01.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 08/22/2014] [Accepted: 01/25/2015] [Indexed: 10/24/2022]
Abstract
N-(2-[(18)F]fluoropropionyl)-l-glutamate([(18)F]FPGLU) was a recently developed potential amino acid tracer for tumor imaging with positron emission tomography-computer tomography (PET-CT). The absorbed and effective radiation doses resulting from the intravenous administration of [(18)F]FPGLU were estimated using biodistribution data from normal mice. The methodology recommended by Medical Internal Radiation Dose Committee (MIRD) was used to estimate the doses. The highest uptake of [(18)F]FPGLU was found in the kidneys, followed by the liver and lung. The kidneys were the organ received the highest absorbed dose, 58.4μGy/MBq, the brain received the lowest dose, 5.5μGy/MBq, and other organs received doses in the range of 8.3-11.9μGy/MBq. The effective dose was 17.0μSv/MBq. The data show that a 370MBq (10mCi) injection of [(18)F]FPGLU would lead to an estimated effective dose of 6.3mSv, which is within the accepted range of routine nuclear medicine investigations.
Collapse
Affiliation(s)
- Caihua Tang
- PET-CT Center, Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China
| | - Zeqing Xu
- Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China
| | - Kongzhen Hu
- PET-CT Center, Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Baoguo Yao
- PET-CT Center, Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Ganghua Tang
- PET-CT Center, Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Dahong Nie
- PET-CT Center, Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| |
Collapse
|
37
|
System L amino acid transporter LAT1 accumulates O-(2-fluoroethyl)-L-tyrosine (FET). Amino Acids 2014; 47:335-44. [PMID: 25385314 DOI: 10.1007/s00726-014-1863-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 10/24/2014] [Indexed: 01/04/2023]
Abstract
O-(2-fluoroethyl)-L-tyrosine (FET) labeled with fluorine-18 is an important and specific tracer for diagnostics of glioblastoma via positron emission tomography (PET). However, the mechanism of its quite specific accumulation in tumor tissue has not been understood so far. In this work we demonstrate that [(3)H]L-tyrosine is primarily transported by the system L transporter LAT1 in human LN229 glioblastoma cells. FET reduced tyrosine transport, suggesting that it shares the same uptake pathway. More importantly, accumulation of FET was significantly reduced after siRNA-mediated downregulation of LAT1. Xenopus laevis oocytes expressing human LAT1 together with the glycoprotein 4F2hc (necessary to pull LAT-1 to the plasma membrane) exhibited a similar accumulation of FET as observed in glioblastoma cells. In contrast, no accumulation was observed in control oocytes, not overexpressing an exogenous transporter. Because LAT1 works exclusively as an exchanger of amino acids, substrates at one side of the membrane stimulate exchange against substrates at the other side. Extracellular FET stimulated the efflux of intracellular [(3)H]L-leucine, demonstrating that FET is indeed an influx substrate for LAT1. However, FET injected into oocytes was not able to stimulate uptake of extracellular [(3)H]L-leucine, indicating that FET is not a good efflux substrate. Our data, therefore, suggest that FET is trapped within cells due to the asymmetry of its intra- and extracellular recognition by LAT1. If also found for other transporters in tumor cells, asymmetric substrate recognition may be further exploited for tumor-specific accumulation of PET-tracers and/or other tumor-related drugs.
Collapse
|
38
|
Lapa C, Linsenmann T, Monoranu CM, Samnick S, Buck AK, Bluemel C, Czernin J, Kessler AF, Homola GA, Ernestus RI, Löhr M, Herrmann K. Comparison of the amino acid tracers 18F-FET and 18F-DOPA in high-grade glioma patients. J Nucl Med 2014; 55:1611-6. [PMID: 25125481 DOI: 10.2967/jnumed.114.140608] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED High-grade gliomas (HGGs) are the most common malignant primary tumors of the central nervous system. PET probes of amino acid transport such as O-(2-(18)F-fluoroethyl)-l-tyrosine ((18)F-FET), 3,4-dihydroxy-6-(18)F-fluoro-l-phenylalanine ((18)F-DOPA), and (11)C-methionine ((11)C-MET) detect primary and recurrent tumors with a high accuracy. (18)F-FET is predominantly used in Europe, whereas amino acid transport imaging is infrequently done in the United States. The aim of this study was to determine whether (18)F-FET and (18)F-DOPA PET/CT provide comparable information in HGG. METHODS Thirty (18)F-FET and (18)F-DOPA PET/CT scans were obtained before surgery or biopsy in 27 patients with high clinical suspicion for primary or recurrent HGG (5 primary, 22 recurrent tumors). (18)F-FET and (18)F-DOPA PET/CT images were compared visually and semiquantitatively (maximum standardized uptake value [SUV(max)], mean SUV [SUV(mean)]). Background (SUV(max) and SUV(mean)) and tumor-to-background ratios (TBRs) were calculated for both PET probes. The degree of (18)F-DOPA uptake in the basal ganglia (SUV(mean)) was also assessed. RESULTS Visual analysis revealed no difference in tumor uptake pattern between the 2 PET probes. The SUV(mean) and SUV(max) for (18)F-FET were higher than those of (18)F-DOPA (4.0 ± 2.0 and 4.9 ± 2.3 vs. 3.5 ± 1.6 and 4.3 ± 2.0, respectively; all P < 0.001). TBRs for SUV(mean) but not for SUV(max) were significantly higher for (18)F-FET than (18)F-DOPA (TBR SUV(mean): 3.8 ± 1.7 vs. 3.4 ± 1.2, P = 0.004; TBR SUV(max): 3.3 ± 1.6 and 3.0 ± 1.1, respectively; P = 0.086). (18)F-DOPA uptake by the basal ganglia was present (SUV(mean), 2.6 ± 0.7) but did not affect tumor visualization. CONCLUSION Whereas visual analysis revealed no significant differences in uptake pattern for (18)F-FET and (18)F-DOPA in patients with primary or recurrent HGG, both SUVs and TBRs for SUV(mean) were significantly higher for (18)F-FET. However, regarding tumor delineation, both tracers performed equally well and seem equally feasible for imaging of primary and recurrent HGG. These findings suggest that both PET probes can be used based on availability in multicenter trials.
Collapse
Affiliation(s)
- Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Thomas Linsenmann
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | - Camelia Maria Monoranu
- Department of Neuropathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Samuel Samnick
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Christina Bluemel
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Johannes Czernin
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California; and
| | - Almuth F Kessler
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | - Gyoergy A Homola
- Department of Neuroradiology, University Hospital Würzburg, Würzburg, Germany
| | - Ralf-Ingo Ernestus
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | - Mario Löhr
- Department of Neurosurgery, University Hospital Würzburg, Würzburg, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California; and
| |
Collapse
|
39
|
Dunet V, Maeder P, Nicod-Lalonde M, Lhermitte B, Pollo C, Bloch J, Stupp R, Meuli R, Prior JO. Combination of MRI and dynamic FET PET for initial glioma grading. Nuklearmedizin 2014; 53:155-61. [PMID: 24737132 DOI: 10.3413/nukmed-0650-14-03] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 04/08/2014] [Indexed: 12/13/2022]
Abstract
AIM MRI and PET with 18F-fluoro-ethyl-tyrosine (FET) have been increasingly used to evaluate patients with gliomas. Our purpose was to assess the additive value of MR spectroscopy (MRS), diffusion imaging and dynamic FET-PET for glioma grading. PATIENTS, METHODS 38 patients (42 ± 15 aged, F/M: 0.46) with untreated histologically proven brain gliomas were included. All underwent conventional MRI, MRS, diffusion sequences, and FET-PET within 3±4 weeks. Performances of tumour FET time-activity-curve, early-to-middle SUVmax ratio, choline / creatine ratio and ADC histogram distribution pattern for gliomas grading were assessed, as compared to histology. Combination of these parameters and respective odds were also evaluated. RESULTS Tumour time-activity-curve reached the best accuracy (67%) when taken alone to distinguish between low and high-grade gliomas, followed by ADC histogram analysis (65%). Combination of time-activity-curve and ADC histogram analysis improved the sensitivity from 67% to 86% and the specificity from 63-67% to 100% (p < 0.008). On multivariate logistic regression analysis, negative slope of the tumour FET time-activity-curve however remains the best predictor of high-grade glioma (odds 7.6, SE 6.8, p = 0.022). CONCLUSION Combination of dynamic FET-PET and diffusion MRI reached good performance for gliomas grading. The use of FET-PET/MR may be highly relevant in the initial assessment of primary brain tumours.
Collapse
Affiliation(s)
- V Dunet
- Vincent Dunet, MD, BSc, Department of Radiology, Lausanne University Hospital, Rue du Bugnon 46, 1011, Lausanne Switzerland, Tel. +41/795 56 05 68, Fax +41/213 14 43 49, E-mail:
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
He S, Tang G, Hu K, Wang H, Wang S, Huang T, Liang X, Tang X. Radiosynthesis and biological evaluation of 5-(3-[18F]fluoropropyloxy)-L-tryptophan for tumor PET imaging. Nucl Med Biol 2013; 40:801-807. [PMID: 23791401 DOI: 10.1016/j.nucmedbio.2013.04.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 04/24/2013] [Accepted: 04/29/2013] [Indexed: 12/16/2022]
Abstract
INTRODUCTION [(18)F]FDG PET has difficulty distinguishing tumor from inflammation in the clinic because of the same high uptake in nonmalignant and inflammatory tissue. In contrast, amino acid tracers do not accumulate in inflamed tissues and thus provide an excellent opportunity for their use in clinical cancer imaging. In this study, we developed a new amino acid tracer 5-(3-[(18)F]Fluoropropyloxy)-L-tryptophan ([(18)F]-L-FPTP) by two-step reactions and performed its biologic evaluation. METHODS [(18)F]-L-FPTP was prepared by [(18)F]fluoropropylation of 5-hydroxy-L-tryptophan disodium salt and purification on C18 cartridges. The biodistribution of [(18)F]-L-FPTP was determined in normal mice and the incorporation of [(18)F]-L-FPTP into tissue proteins was investigated. In vitro competitive inhibition experiments were performed with Hepa1-6 hepatoma cell lines. [(18)F]-L-FPTP PET imaging was performed on tumor-bearing and inflammation mice and compared with [(18)F]-L-FEHTP PET. RESULTS The overall uncorrected radiochemical yield of [(18)F]-L-FPTP was 21.1 ± 4.4% with a synthesis time of 60 min, the radiochemical purity was more than 99%. Biodistribution studies demonstrate high uptake of [(18)F]-L-FPTP in liver, kidney, pancreas, and blood at the early phase, and fast clearance in most tissues over the whole observed time. The uptake studies in Hepa1-6 cells suggest that [(18)F]-L-FPTP is transported by the amino acid transport system B(0,+), LAT2 and ASC. [(18)F]-L-FPTP displays good stability and is not incorporated into proteins in vitro. PET imaging shows that [(18)F]-L-FPTP can be a better potential PET tracer for differentiating tumor from inflammation than [(18)F]FDG and 5-(3-[(18)F]fluoroethyloxy)-L-tryptophan ([(18)F]-L-FEHTP), with high [(18)F]-L-FPTP uptake ratio (2.53) of tumor to inflammation at 60 min postinjection. CONCLUSIONS Using [(18)F]fluoropropyl derivatives as intermediates, the new tracer [(18)F]-L-FPTP was achieved with good yield and radiochemical purity, and the biological evaluation results of [(18)F]-L-FPTP showed that it was a hopeful tracer for PET tumor imaging.
Collapse
Affiliation(s)
- Shanzhen He
- PET-CT center, Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Smolarz K, Krause BJ, Graner FP, Wagner FM, Wester HJ, Sell T, Bacher-Stier C, Fels L, Dinkelborg L, Schwaiger M. Biodistribution and radiation dosimetry in healthy volunteers of a novel tumour-specific probe for PET/CT imaging: BAY 85-8050. Eur J Nucl Med Mol Imaging 2013; 40:1861-8. [PMID: 23877631 DOI: 10.1007/s00259-013-2502-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 06/28/2013] [Indexed: 12/25/2022]
Abstract
PURPOSE Novel tracers for the diagnosis of malignant disease with PET and PET/CT are being developed as the most commonly used (18)F deoxyglucose (FDG) tracer shows certain limitations. Employing radioactively labelled glutamate derivatives for specific imaging of the truncated citrate cycle potentially allows more specific tumour imaging. Radiation dosimetry of the novel tracer BAY 85-8050, a glutamate derivative, was calculated and the effective dose (ED) was compared with that of FDG. METHODS Five healthy volunteers were included in the study. Attenuation-corrected whole-body PET/CT scans were performed from 0 to 90 min, at 120 and at 240 min after injection of 305.0 ± 17.6 MBq of BAY 85-8050. Organs with moderate to high uptake at any of the imaging time points were used as source organs. Total activity in each organ at each time point was measured. Time-activity curves (TAC) were determined for the whole body and all source organs. The resulting TACs were fitted to exponential equations and accumulated activities were determined. OLINDA/EXM software was used to calculate individual organ doses and the whole-body ED from the acquired data. RESULTS Uptake of the tracer was highest in the kidneys due to renal excretion of the tracer, followed by the pancreas, heart wall and osteogenic cells. The mean organ doses were: kidneys 38.4 ± 11.2 μSv/MBq, pancreas 23.2 ± 3.8 μSv/MBq, heart wall 17.4 ± 4.1 μSv/MBq, and osteogenic cells 13.6 ± 3.5 μSv/MBq. The calculated ED was 8.9 ± 1.5 μSv/MBq. CONCLUSION Based on the distribution and dose estimates, the calculated radiation dose of BAY 85-8050 is 2.67 ± 0.45 mSv at a patient dose of 300 MBq, which compares favourably with the radiation dose of FDG (5.7 mSv).
Collapse
Affiliation(s)
- Kamilla Smolarz
- Department of Nuclear Medicine, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Smolarz K, Krause BJ, Graner FP, Wagner FM, Hultsch C, Bacher-Stier C, Sparks RB, Ramsay S, Fels LM, Dinkelborg LM, Schwaiger M. (S)-4-(3-18F-Fluoropropyl)-l-Glutamic Acid: An 18F-Labeled Tumor-Specific Probe for PET/CT Imaging—Dosimetry. J Nucl Med 2013; 54:861-6. [DOI: 10.2967/jnumed.112.112581] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
43
|
Diagnostic accuracy of F-18-fluoroethyltyrosine PET and PET/CT in patients with brain tumor. Clin Transl Imaging 2013. [DOI: 10.1007/s40336-013-0017-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
44
|
|
45
|
Radiosynthesis and biological evaluation of alpha-[F-18]fluoromethyl phenylalanine for brain tumor imaging. Nucl Med Biol 2013; 40:498-506. [PMID: 23528560 DOI: 10.1016/j.nucmedbio.2012.12.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 11/19/2012] [Accepted: 12/10/2012] [Indexed: 11/20/2022]
Abstract
OBJECTIVES Radiolabeled amino acids have proven utility for imaging brain tumors in humans, particularly those that target system L amino acid transport. We have prepared the novel phenylalanine analogue, (FMePhe, 9), as part of an effort to develop new system L tracers that can be prepared in high radiochemical yield through nucleophilic [(18)F]fluorination. The tumor imaging properties of both enantiomers of this new tracer were evaluated through cell uptake, biodistribution and microPET studies in the mouse DBT model of high grade glioma. METHODS The non-radioactive form of 9 and the cyclic sulfamidate labeling precursor were prepared from commercially available racemic α-benzylserine. Racemic [(18)F]9 was prepared from the labeling precursor in two steps using standard[(18)F]fluoride nucleophilic reaction conditions followed by acidic deprotection. The individual enantiomers [(18)F]9a and [(18)F]9b were isolated using preparative chiral HPLC. In vitro uptake inhibition assays were performed with each enantiomer using DBT cells. Biodistribution and microPET/CT studies were performed with each enantiomer in male BALB/c mice at approximately 2 weeks after implantation of DBT tumor cells. RESULTS Radiolabeling of the cyclic sulfamidate precursor 5 provides racemic [(18)F]9 in high radiochemical yield (60%-70%, n=4) and high radiochemical purity (>96%, n=4). In vitro uptake assays demonstrate that both [(18)F]9a and [(18)F]9b undergo tumor cell uptake through system L transport. The biodistribution studies using the single enantiomers [(18)F]9a and [(18)F]9b demonstrated good tumor uptake with lower uptake in most normal tissues, and [(18)F]9a had higher tumor uptake than [(18)F]9b. MicroPET imaging demonstrated good tumor visualization within 10 min of injection, rapid uptake of radioactivity, and tumor to brain ratios of approximately 6:1 at 60 min postinjection. CONCLUSIONS The novel PET tracer, [(18)F]FMePhe, is readily synthesized in good yield from a cyclic sulfamidate precursor. Biodistribution and microPET studies in the DBT model demonstrate good tumor to tissue ratios and tumor visualization, with enantiomer [(18)F]9a having higher tumor uptake. However, the brain availability of both enantiomers was lower than expected for system L substrates, suggesting the [(18)F]fluorine group in the β-position affects uptake of these compounds by system L transporters.
Collapse
|
46
|
Sakata M, Oda K, Toyohara J, Ishii K, Nariai T, Ishiwata K. Direct comparison of radiation dosimetry of six PET tracers using human whole-body imaging and murine biodistribution studies. Ann Nucl Med 2013; 27:285-96. [DOI: 10.1007/s12149-013-0685-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 01/06/2013] [Indexed: 11/30/2022]
|
47
|
Franck D, Kniess T, Steinbach J, Zitzmann-Kolbe S, Friebe M, Dinkelborg LM, Graham K. Investigations into the synthesis, radiofluorination and conjugation of a new [¹⁸F]fluorocyclobutyl prosthetic group and its in vitro stability using a tyrosine model system. Bioorg Med Chem 2012; 21:643-52. [PMID: 23290251 DOI: 10.1016/j.bmc.2012.11.049] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 11/12/2012] [Accepted: 11/29/2012] [Indexed: 11/29/2022]
Abstract
The [(18)F]fluorocyclobutyl group has the potential to be a metabolically stable prosthetic group for PET tracers. The synthesis of the radiolabeling precursor cis-cyclobutane-1,3-diyl bis(toluene-4-sulfonate) 8 was obtained from epibromohydrin in 7 steps (2% overall yield). The radiolabeling of this precursor 8 and its conjugation to L-tyrosine as a model system was successfully achieved to give the new non-natural amino acid 3-[(18)F]fluorocyclobutyl-L-tyrosine (L-3-[(18)F]FCBT) [(18)F]17 in 8% decay-corrected yield from the non-carrier-added [(18)F]fluoride. L-3-[(18)F]FCBT was investigated in vitro in different cancer cell lines to determine the uptake and stability. The tracer [(18)F]17 showed a time dependent uptake into different tumor cell lines (A549, NCI-H460, DU145) with the best uptake of 5.8% injected dose per 5×10(5) cells after 30min in human lung carcinoma cells A549. The stability of L-3-[(18)F]FCBT in human and rat plasma and the stability of the non-radioactive L-3-FCBT in rat hepatocytes were both found to be excellent. These results show that the non-natural amino acid L-3-[(18)F]FCBT is a promising metabolically stable radiotracer for positron emission tomography.
Collapse
Affiliation(s)
- Dominic Franck
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | | | | | | | | | | | | |
Collapse
|
48
|
Wiriyasermkul P, Nagamori S, Tominaga H, Oriuchi N, Kaira K, Nakao H, Kitashoji T, Ohgaki R, Tanaka H, Endou H, Endo K, Sakurai H, Kanai Y. Transport of 3-fluoro-L-α-methyl-tyrosine by tumor-upregulated L-type amino acid transporter 1: a cause of the tumor uptake in PET. J Nucl Med 2012; 53:1253-61. [PMID: 22743251 DOI: 10.2967/jnumed.112.103069] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED l-3-(18)F-α-methyl tyrosine ((18)F-FAMT) has been developed as a PET radiotracer for tumor imaging. Clinical studies have demonstrated the usefulness of (18)F-FAMT PET for the prediction of prognosis and the differentiation of malignant tumors and benign lesions. (18)F-FAMT exhibits higher cancer specificity in peripheral organs than other amino acid PET tracers and (18)F-FDG. The accumulation of (18)F-FAMT is strongly correlated with the expression of L-type amino acid transporter 1 (LAT1), an isoform of system L highly upregulated in cancers. In this study, we examined the interaction of 3-fluoro-l-α-methyl-tyrosine (FAMT) with amino acid transporters to assess the mechanisms of (18)F-FAMT uptake in PET. METHODS We applied in vitro assays using established mammalian cell lines stably expressing LAT1 or a non-cancer-type system L isoform LAT2. The inhibitory effect on l-(14)C-leucine uptake and the induction effect on efflux of preloaded l-(14)C-leucine were examined for FAMT and other amino acid tracers. FAMT transport was compared among cell lines with varied LAT1 expression level. RESULTS FAMT prominently inhibited LAT1-mediated l-(14)C-leucine uptake in a competitive manner but had less of an effect on LAT2. In the efflux experiments, FAMT induced the efflux of preloaded l-(14)C-leucine through LAT1, indicating that FAMT is transported by LAT1 and not by LAT2. Among amino acid-related compounds examined in this study, including those used for PET tracers, the compounds with an α-methyl group such as FAMT, 2-fluoro-l-α-methyl-tyrosine, 3-iodo-l-α-methyl-tyrosine, and l-α-methyl-tyrosine were well transported by LAT1 but not by LAT2. However, l-methionine, l-tyrosine, 3-fluoro-l-tyrosine, 2-fluoro-l-tyrosine, and O-(2-fluoroethyl)-l-tyrosine were transported by both LAT1 and LAT2, suggesting that the α-methyl moiety is responsible for the LAT1 selectivity of FAMT. FAMT transport rate and LAT1 protein level were well correlated, supporting the importance of LAT1 for the cellular uptake of FAMT. CONCLUSION Distinct from other amino acid PET tracers, because of its α-methyl moiety, FAMT is selective to LAT1 and not transported by LAT2. This property of FAMT is proposed to contribute to highly tumor-specific accumulation of (18)F-FAMT in PET.
Collapse
Affiliation(s)
- Pattama Wiriyasermkul
- Division of Bio-system Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Heinzel A, Stock S, Langen KJ, Müller D. Cost-effectiveness analysis of amino acid PET-guided surgery for supratentorial high-grade gliomas. J Nucl Med 2012; 53:552-8. [PMID: 22419752 DOI: 10.2967/jnumed.111.097352] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
UNLABELLED High-grade gliomas are brain tumors associated with a devastating prognosis. Recent studies have indicated that the combined use of amino acid PET and MRI is superior to MRI alone to plan the surgical resection of high-grade gliomas. The aim of the study was to analyze the cost-effectiveness of the use of amino acid PET for the surgical resection of high-grade gliomas, compared with MRI alone, from the perspective of the national health insurance in Germany. METHODS A decision-tree model was set up to compare 2 strategies: the use of MRI alone and the combined use of MRI and PET for surgical resection of high-grade gliomas. For the analysis, 2 scenarios were calculated: a baseline scenario and a more expensive scenario, accounting for disease severity. To test the robustness of the results, probabilistic sensitivity analyses using Monte Carlo simulation were calculated. RESULTS Compared with MRI alone, the combined use of MRI and PET showed an incremental cost-effectiveness ratio of €2,948 (€1 ~ U.S.$1.3)per life-year gained for the baseline scenario and an incremental cost-effectiveness ratio of €4,105 per life-year gained for the admissible-charge-rate scenario. In the probabilistic sensitivity analysis in about 60% of the iterations, the combined use of PET and MRI was superior to MRI alone when assuming a willingness-to-pay threshold of €30,000. CONCLUSION The model indicates that the combined use of MRI and PET may be cost-effective. The results of this analysis have to be considered carefully because there was only limited empiric evidence for several input parameters.
Collapse
Affiliation(s)
- Alexander Heinzel
- Department of Nuclear Medicine of the Heinrich-Heine University of Duesseldorf at the Research Centre Juelich, Juelich, Germany.
| | | | | | | |
Collapse
|
50
|
Tavares AAS, Lewsey J, Dewar D, Pimlott SL. Radiotracer properties determined by high performance liquid chromatography: a potential tool for brain radiotracer discovery. Nucl Med Biol 2012; 39:127-35. [DOI: 10.1016/j.nucmedbio.2011.06.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 06/17/2011] [Accepted: 06/27/2011] [Indexed: 01/14/2023]
|