1
|
Sharkey AR, Koglin N, Mittra ES, Han S, Cook GJR, Witney TH. Clinical [ 18F]FSPG Positron Emission Tomography Imaging Reveals Heterogeneity in Tumor-Associated System x c- Activity. Cancers (Basel) 2024; 16:1437. [PMID: 38611114 PMCID: PMC11011143 DOI: 10.3390/cancers16071437] [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: 02/09/2024] [Revised: 03/31/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND (4S)-4-(3-[18F]fluoropropyl)-L-glutamic acid ([18F]FSPG) positron emission tomography/computed tomography (PET/CT) provides a readout of system xc- transport activity and has been used for cancer detection in clinical studies of different cancer types. As system xc- provides the rate-limiting precursor for glutathione biosynthesis, an abundant antioxidant, [18F]FSPG imaging may additionally provide important prognostic information. Here, we performed an analysis of [18F]FSPG radiotracer distribution between primary tumors, metastases, and normal organs from cancer patients. We further assessed the heterogeneity of [18F]FSPG retention between cancer types, and between and within individuals. METHODS This retrospective analysis of prospectively collected data compared [18F]FSPG PET/CT in subjects with head and neck squamous cell cancer (HNSCC, n = 5) and non-small-cell lung cancer (NSCLC, n = 10), scanned at different institutions. Using semi-automated regions of interest drawn around tumors and metastases, the maximum standardized uptake value (SUVmax), SUVmean, SUV standard deviation and SUVpeak were measured. [18F]FSPG time-activity curves (TACs) for normal organs, primary tumors and metastases were subsequently compared to 18F-2-fluoro-2-deoxy-D-glucose ([18F]FDG) PET/CT at 60 min post injection (p.i.). RESULTS The mean administered activity of [18F]FSPG was 309.3 ± 9.1 MBq in subjects with NSCLC and 285.1 ± 11.3 MBq in those with HNSCC. The biodistribution of [18F]FSPG in both cohorts showed similar TACs in healthy organs from cancer patients. There was no statistically significant overall difference in the average SUVmax of tumor lesions at 60 min p.i. for NSCLC (8.1 ± 7.1) compared to HNSCC (6.0 ± 4.1; p = 0.29) for [18F]FSPG. However, there was heterogeneous retention between and within cancer types; the SUVmax at 60 min p.i. ranged from 1.4 to 23.7 in NSCLC and 3.1-12.1 in HNSCC. CONCLUSION [18F]FSPG PET/CT imaging from both NSCLC and HNSCC cohorts showed the same normal-tissue biodistribution, but marked tumor heterogeneity across subjects and between lesions. Despite rapid elimination through the urinary tract and low normal-background tissue retention, the diagnostic potential of [18F]FSPG was limited by variability in tumor retention. As [18F]FSPG retention is mediated by the tumor's antioxidant capacity and response to oxidative stress, this heterogeneity may provide important insights into an individual tumor's response or resistance to therapy.
Collapse
Affiliation(s)
- Amy R. Sharkey
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London SE1 7EH, UK; (A.R.S.); (G.J.R.C.)
| | | | - Erik S. Mittra
- Division of Molecular Imaging and Therapy, Oregon Health & Science University, Portland, OR 97239, USA;
| | - Sangwon Han
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea;
| | - Gary J. R. Cook
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London SE1 7EH, UK; (A.R.S.); (G.J.R.C.)
- King’s College London and Guy’s and St. Thomas’ PET Center, St. Thomas’ Hospital, London SE1 7EH, UK
| | - Timothy H. Witney
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London SE1 7EH, UK; (A.R.S.); (G.J.R.C.)
| |
Collapse
|
2
|
Constantinescu CC, Brown T, Wang S, Yin W, Barret O, Jennings D, Tauscher J. Clinical Characterization of [ 18F]T-008, a Cholesterol 24-Hydroxylase PET Ligand: Dosimetry, Kinetic Modeling, Variability, and Soticlestat Occupancy. J Nucl Med 2023; 64:1972-1979. [PMID: 37770111 PMCID: PMC10690114 DOI: 10.2967/jnumed.123.265912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/18/2023] [Indexed: 10/03/2023] Open
Abstract
This series of studies characterized [18F]T-008, a PET radiotracer for imaging cholesterol 24-hydroxylase (CH24H), in healthy volunteers (ClinicalTrials.gov identifier NCT02497235). Assessments included radiation dosimetry, kinetic modeling, test-retest variability (TRT) evaluation, and a dose occupancy evaluation using soticlestat, a selective CH24H inhibitor. Soticlestat is currently in phase 3 development for the treatment of seizures in Dravet syndrome and Lennox-Gastaut syndrome. Methods: In the dosimetry study, 5 participants (3 men) underwent serial whole-body scans to estimate organ-absorbed doses and effective doses of [18F]T-008 using OLINDA/EXM 1.1. For the kinetic modeling and TRT study, 6 participants (all men) underwent two 210-min dynamic [18F]T-008 PET scans with arterial blood sampling. The regional total volume of distribution was estimated using a 1-tissue-compartment model, a 2-tissue-compartment model, and Logan graphic analysis. In the dose occupancy study, 11 participants (all men) underwent 120-min scans at baseline and 2 time points (peak and trough) after receiving single oral doses of soticlestat (50-600 mg). The relationship between effect-site soticlestat concentration and brain occupancy was evaluated with a specially developed pharmacokinetic model and a saturable maximal occupancy model. Results: The estimated mean whole-body effective dose was 0.0292 mSv/MBq (SD, 0.00147 mSv/MBq). [18F]T-008 entered the brain rapidly, with a distribution consistent with known CH24H distribution densities. The 2-tissue-compartment model and Logan graphic analysis best described the tracer kinetics. The mean TRT for estimating total volume of distribution was 7%-15%. Single doses of soticlestat in the range 50-600 mg resulted in occupancies of 64%-96% at 2 h and 11%-79% at 24 h. The estimated half-maximal effect-site concentration of soticlestat was 5.52 ng/mL. Conclusion: [18F]T-008 is a suitable PET radiotracer for quantitatively analyzing CH24H in the human brain. Using [18F]T-008 and PET, we demonstrated that soticlestat was brain-penetrant and established target engagement by displacing [18F]T-008 in a dose-dependent manner in the brain.
Collapse
Affiliation(s)
| | - Terry Brown
- Takeda Pharmaceutical Co. Ltd., Cambridge, Massachusetts
| | - Shining Wang
- Takeda Pharmaceutical Co. Ltd., Cambridge, Massachusetts
| | - Wei Yin
- Takeda Pharmaceutical Co. Ltd., Cambridge, Massachusetts
| | | | | | | |
Collapse
|
3
|
Sharkey AR, Witney TH, Cook GJR. Is System x c- a Suitable Target for Tumour Detection and Response Assessment with Imaging? Cancers (Basel) 2023; 15:5573. [PMID: 38067277 PMCID: PMC10705217 DOI: 10.3390/cancers15235573] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 02/12/2024] Open
Abstract
System xc- is upregulated in cancer cells and can be imaged using novel radiotracers, most commonly with (4S)-4-(3-[18F]fluoropropyl)-L-glutamic acid (18F-FSPG). The aim of this review was to summarise the use of 18F-FSPG in humans, explore the benefits and limitations of 18F-FSPG, and assess the potential for further use of 18F-FSPG in cancer patients. To date, ten papers have described the use of 18F-FSPG in human cancers. These studies involved small numbers of patients (range 1-26) and assessed the use of 18F-FSPG as a general oncological diagnostic agent across different cancer types. These clinical trials were contrasting in their findings, limiting the scope of 18F-FSPG PET/CT as a purely diagnostic agent, primarily due to heterogeneity of 18F-FSPG retention both between cancer types and patients. Despite these limitations, a potential further application for 18F-FSPG is in the assessment of early treatment response and prediction of treatment resistance. Animal models of cancer have shown that changes in 18F-FSPG retention following effective therapy precede glycolytic changes, as indicated by 18F-FDG, and changes in tumour volume, as measured by CT. If these results could be replicated in human clinical trials, imaging with 18F-FSPG PET/CT would offer an exciting route towards addressing the currently unmet clinical needs of treatment resistance prediction and early imaging assessment of therapy response.
Collapse
Affiliation(s)
- Amy R. Sharkey
- School of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, UK
| | - Timothy H. Witney
- School of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, UK
| | - Gary J. R. Cook
- School of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, UK
- King’s College London and Guy’s and St. Thomas’ PET Centre, St. Thomas’ Hospital, London SE1 7EH, UK
| |
Collapse
|
4
|
Calatayud DG, Lledos M, Casarsa F, Pascu SI. Functional Diversity in Radiolabeled Nanoceramics and Related Biomaterials for the Multimodal Imaging of Tumors. ACS BIO & MED CHEM AU 2023; 3:389-417. [PMID: 37876497 PMCID: PMC10591303 DOI: 10.1021/acsbiomedchemau.3c00021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/18/2023] [Accepted: 07/18/2023] [Indexed: 10/26/2023]
Abstract
Nanotechnology advances have the potential to assist toward the earlier detection of diseases, giving increased accuracy for diagnosis and helping to personalize treatments, especially in the case of noncommunicative diseases (NCDs) such as cancer. The main advantage of nanoparticles, the scaffolds underpinning nanomedicine, is their potential to present multifunctionality: synthetic nanoplatforms for nanomedicines can be tailored to support a range of biomedical imaging modalities of relevance for clinical practice, such as, for example, optical imaging, computed tomography (CT), magnetic resonance imaging (MRI), single photon emission computed tomography (SPECT), and positron emission tomography (PET). A single nanoparticle has the potential to incorporate myriads of contrast agent units or imaging tracers, encapsulate, and/or be conjugated to different combinations of imaging tags, thus providing the means for multimodality diagnostic methods. These arrangements have been shown to provide significant improvements to the signal-to-noise ratios that may be obtained by molecular imaging techniques, for example, in PET diagnostic imaging with nanomaterials versus the cases when molecular species are involved as radiotracers. We surveyed some of the main discoveries in the simultaneous incorporation of nanoparticulate materials and imaging agents within highly kinetically stable radio-nanomaterials as potential tracers with (pre)clinical potential. Diversity in function and new developments toward synthesis, radiolabeling, and microscopy investigations are explored, and preclinical applications in molecular imaging are highlighted. The emphasis is on the biocompatible materials at the forefront of the main preclinical developments, e.g., nanoceramics and liposome-based constructs, which have driven the evolution of diagnostic radio-nanomedicines over the past decade.
Collapse
Affiliation(s)
- David G. Calatayud
- Department
of Inorganic Chemistry, Universidad Autónoma
de Madrid, Madrid 28049, Spain
- Department
of Electroceramics, Instituto de Cerámica
y Vidrio, Madrid 28049, Spain
| | - Marina Lledos
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Federico Casarsa
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Sofia I. Pascu
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
- Centre
of Therapeutic Innovations, University of
Bath, Bath BA2 7AY, United Kingdom
| |
Collapse
|
5
|
Huang Y, Liu Y, Li C, Li Z, Chen H, Zhang L, Liang Y, Wu Z. Evaluation of (2S,4S)-4-[ 18F]FEBGln as a Positron Emission Tomography Tracer for Tumor Imaging. Mol Pharm 2023; 20:5195-5205. [PMID: 37647563 DOI: 10.1021/acs.molpharmaceut.3c00544] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Glutamine metabolism-related tracers have the potential to visualize numerous tumors because glutamine is the second largest source of energy for tumors. (2S,4S)-4-[18F]FEBGln was designed by introducing [18F]fluoroethoxy benzyl on carbon-4 of glutamine. The aim of this study was to investigate the pharmacokinetic properties and tumor positron emission tomography (PET) imaging characteristics of (2S,4S)-4-[18F]FEBGln in detail. The biodistribution results of nude mice bearing MCF-7 tumor showed that (2S,4S)-4-[18F]FEBGln had high initial tumor uptake, and a fast clearance rate, resulting in a high tumor-to-muscle ratio at 30 min postinjection. There was no obvious defluorination in vivo. The micro-PET-CT imaging results of (2S,4S)-4-[18F]FEBGln orthotopic MCF-7 tumor-bearing nude mice were consistent with the biological distribution results. Compared with (2S,4R)-4-[18F]FGln, (2S,4S)-4-[18F]FEBGln showed poor tumor retention, but its clearance in normal tissues was also fast, so it had better PET image contrast than the former. Unlike poor retention in MCF-7-bearing nude mice, (2S,4S)-4-[18F]FEBGln has good retention in NCI-h1975 and 22Rv1 tumor models. Since (2S,4S)-4-[18F]FEBGln has low uptake in normal lungs and high uptake in the bladder, it is expected to be used in the accurate diagnosis of lung cancer but cannot accurately determine prostate cancer. Consistent with the advantages of radiolabeled amino acids in the application of brain tumors, (2S,4S)-4-[18F]FEBGln accurately diagnoses U87MG glioma with higher contrast than [18F]FET and [18F]FDG, and there is a correlation between (2S,4S)-4-[18F]FEBGln uptake and tumor growth cycle. Further kinetic model analysis showed that (2S,4S)-4-[18F]FEBGln was similar to (2S,4R)-4-[18F]FGln, conforming to the one-compartment model and the Logan graphical model, and was expected to assess the size of the glutamine pool of the tumor. Therefore, (2S,4S)-4-[18F]FEBGln is expected to provide a strong imaging basis for the diagnosis, formulation of personalized plans, and efficacy evaluation of glioma, lung cancer, and breast cancer.
Collapse
Affiliation(s)
- Yong Huang
- Department of Nuclear Medicine, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Yajing Liu
- School of Pharmaceutical Science, Capital Medical University, Beijing 100069, China
| | - Chengze Li
- Department of Nuclear Medicine, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Zhongjing Li
- Department of Nuclear Medicine, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Hualong Chen
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Lu Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Ying Liang
- Department of Nuclear Medicine, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Zehui Wu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| |
Collapse
|
6
|
Lin M, Ta RT, Manning HC. Simplified and highly-reliable automated production of [ 18F]FSPG for clinical studies. EJNMMI Radiopharm Chem 2023; 8:15. [PMID: 37486582 PMCID: PMC10366059 DOI: 10.1186/s41181-023-00200-8] [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: 06/13/2023] [Accepted: 07/13/2023] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND (S)-4-(3-18F-Fluoropropyl)-L-Glutamic Acid ([18F]FSPG) is a positron emission tomography (PET) tracer that specifically targets the cystine/glutamate antiporter (xc-), which is frequently overexpressed in cancer and several neurological disorders. Pilot studies examining the dosimetry and biodistribution of [18F]FSPG in healthy volunteers and tumor detection in patients with non-small cell lung cancer, hepatocellular carcinoma, and brain tumors showed promising results. In particular, low background uptake in the brain, lung, liver, and bowel was observed that further leads to excellent imaging contrasts of [18F]FSPG PET. However, reliable production-scale cGMP-compliant automated procedures for [18F]FSPG production are still lacking to further increase the utility and clinical adoption of this radiotracer. Herein, we report the optimized automated approaches to produce [18F]FSPG through two commercially available radiosynthesizers capable of supporting centralized and large-scale production for clinical use. RESULTS Starting with activity levels of 60-85 GBq, the fully-automated process to produce [18F]FSPG took less than 45 min with average radiochemical yields of 22.56 ± 0.97% and 30.82 ± 1.60% (non-decay corrected) using TRACERlab™ FXFN and FASTlab™, respectively. The radiochemical purities were > 95% and the formulated [18F]FSPG solution was determined to be sterile and colorless with the pH of 6.5-7.5. No radiolysis of the product was observed up to 8 h after final batch formulation. CONCLUSIONS In summary, cGMP-compliant radiosyntheses and quality control of [18F]FSPG have been established on two commercially available synthesizers leveraging high activity concentration and radiochemical purity. While the clinical trials using [18F]FSPG PET are currently underway, the automated approaches reported herein will accelerate the clinical adoption of this radiotracer and warrant centralized and large-scale production of [18F]FSPG.
Collapse
Affiliation(s)
- Mai Lin
- Cyclotron Radiochemistry Facility, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Robert T Ta
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - H Charles Manning
- Cyclotron Radiochemistry Facility, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA.
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
7
|
Lin M, Ta RT, Charles Manning H. Simplified and Highly-reliable automated production of [18F]FSPG for clinical studies. RESEARCH SQUARE 2023:rs.3.rs-3031030. [PMID: 37461634 PMCID: PMC10350228 DOI: 10.21203/rs.3.rs-3031030/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2023]
Abstract
Background (S)-4-(3- 18 F-Fluoropropyl)-L-Glutamic Acid ([ 18 F]FSPG) is a positron emission tomography (PET) tracer that specifically targets the cystine/glutamate antiporter (xc-), which is frequently overexpressed in cancer and several neurological disorders. Pilot studies examining the dosimetry and biodistribution of ([ 18 F]FSPG in healthy volunteers and tumor detection in patients with non-small cell lung cancer, hepatocellular carcinoma, and brain tumors showed promising results. In particular, low background uptake in the brain, lung, liver, and bowel was observed that further leads to excellent imaging contrasts of [ 18 F]FSPG PET. However, reliable production-scale cGMP-compliant automated procedures for [ 18 F]FSPG production are still lacking to further increase the utility and clinical adoption of this radiotracer. Herein, we report the optimized automated approaches to produce [ 18 F]FSPG through two commercially available radiosynthesizers capable of supporting centralized and large-scale production for clinical use. Results Starting with activity levels of 60-85 GBq, the fully-automated process to produce [ 18 F]FSPG took less than 45 minutes with average radiochemical yields of 22.56 ± 0.97% and 30.82 ± 1.60% (non-decay corrected) using TRACERlab™ FXFN and FASTlab™, respectively. The radiochemical purities were > 95% and the formulated [ 18 F]FSPG solution was determined to be sterile and colorless with the pH of 6.5-7.5. No radiolysis of the product was observed up to 8 hours after final batch formulation. Conclusions In summary, cGMP-compliant radiosyntheses and quality control of [ 18 F]FSPG have been established on two commercially available synthesizers leveraging high activity concentration and radiochemical purity. While the clinical trials using [ 18 F]FSPG PET are currently underway, the automated approaches reported herein will accelerate the clinical adoption of this radiotracer and warrant centralized and large-scale production of [ 18 F]FSPG.
Collapse
Affiliation(s)
- Mai Lin
- The University of Texas MD Anderson Cancer Center
| | - Robert T Ta
- The University of Texas MD Anderson Cancer Center
| | | |
Collapse
|
8
|
Brown G, Soloviev D, Lewis DY. Radiosynthesis and Analysis of (S)-4-(3-[ 18F]Fluoropropyl)-L-Glutamic Acid. Mol Imaging Biol 2023; 25:586-595. [PMID: 36525163 PMCID: PMC10172245 DOI: 10.1007/s11307-022-01793-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/23/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022]
Abstract
PURPOSE (S)-4-(3-[18F]Fluoropropyl)-L-glutamic acid ([18F]FSPG) is an L-glutamate derivative used as a PET biomarker to assess intracellular redox status in vivo through targeting of the cystine/glutamate antiporter protein, xc- transporter. In this report, we describe a radiosynthesis of [18F]FSPG for use in PET studies that address specific challenges in relation to the radiotracer purity, molar activity, and quality control testing methods. PROCEDURES The radiosynthesis of [18F]FSPG was performed using a customised RNPlus Research automated radiosynthesis system (Synthra GmbH, Hamburg, Germany). [18F]FSPG was labelled in the 3-fluoropropylmoiety at the 4-position of the glutamic acid backbone with fluorine-18 via substitution of nucleophilic [18F]fluoride with a protected naphthylsulfonyloxy-propyl-L-glutamate derivative. Radiochemical purity of the final product was determined by radio HPLC using a new method of direct analysis using a Hypercarb C18 column. RESULTS The average radioactivity yield of [18F]FSPG was 4.2 GBq (range, 3.4-4.8 GBq) at the end of synthesis, starting from 16 GBq of [18F]fluoride at the end of bombardment (n = 10) in a synthesis time of 50 min. The average molar activity and radioactivity volumetric concentration at the end of synthesis were 66 GBq µmol-1 (range, 48-73 GBq µmol-1) and 343-400 MBq mL-1, respectively. CONCLUSION Stability tests using a 4.6 GBq dose with a radioactivity volumetric concentration of 369 MBq mL-1 at the end of synthesis showed no observable radiolysis 3 h after production. The formulated product is of high radiochemical purity (> 95%) and higher molar activity compared to previous methods and is safe to inject into mice up to 3 h after production.
Collapse
Affiliation(s)
- Gavin Brown
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Dmitry Soloviev
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - David Y Lewis
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK.
- School of Cancer Sciences, University of Glasgow, Glasgow, G611QH, UK.
| |
Collapse
|
9
|
Lin M, Coll RP, Cohen AS, Georgiou DK, Manning HC. PET Oncological Radiopharmaceuticals: Current Status and Perspectives. Molecules 2022; 27:6790. [PMID: 36296381 PMCID: PMC9609795 DOI: 10.3390/molecules27206790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/03/2022] [Accepted: 10/07/2022] [Indexed: 02/01/2024] Open
Abstract
Molecular imaging is the visual representation of biological processes that take place at the cellular or molecular level in living organisms. To date, molecular imaging plays an important role in the transition from conventional medical practice to precision medicine. Among all imaging modalities, positron emission tomography (PET) has great advantages in sensitivity and the ability to obtain absolute imaging quantification after corrections for photon attenuation and scattering. Due to the ability to label a host of unique molecules of biological interest, including endogenous, naturally occurring substrates and drug-like compounds, the role of PET has been well established in the field of molecular imaging. In this article, we provide an overview of the recent advances in the development of PET radiopharmaceuticals and their clinical applications in oncology.
Collapse
Affiliation(s)
- Mai Lin
- Cyclotron Radiochemistry Facility, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Ryan P. Coll
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Allison S. Cohen
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dimitra K. Georgiou
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Henry Charles Manning
- Cyclotron Radiochemistry Facility, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
10
|
Zhu J, Pan F, Cai H, Pan L, Li Y, Li L, Li Y, Wu X, Fan H. Positron emission tomography imaging of lung cancer: An overview of alternative positron emission tomography tracers beyond F18 fluorodeoxyglucose. Front Med (Lausanne) 2022; 9:945602. [PMID: 36275809 PMCID: PMC9581209 DOI: 10.3389/fmed.2022.945602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
Lung cancer has been the leading cause of cancer-related mortality in China in recent decades. Positron emission tomography-computer tomography (PET/CT) has been established in the diagnosis of lung cancer. 18F-FDG is the most widely used PET tracer in foci diagnosis, tumor staging, treatment planning, and prognosis assessment by monitoring abnormally exuberant glucose metabolism in tumors. However, with the increasing knowledge on tumor heterogeneity and biological characteristics in lung cancer, a variety of novel radiotracers beyond 18F-FDG for PET imaging have been developed. For example, PET tracers that target cellular proliferation, amino acid metabolism and transportation, tumor hypoxia, angiogenesis, pulmonary NETs and other targets, such as tyrosine kinases and cancer-associated fibroblasts, have been reported, evaluated in animal models or under clinical investigations in recent years and play increasing roles in lung cancer diagnosis. Thus, we perform a comprehensive literature review of the radiopharmaceuticals and recent progress in PET tracers for the study of lung cancer biological characteristics beyond glucose metabolism.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China,Respiratory and Critical Care Medicine, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China,NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Fei Pan
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Huawei Cai
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lili Pan
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yalun Li
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lin Li
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - YunChun Li
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China,Department of Nuclear Medicine, The Second People’s Hospital of Yibin, Yibin, China
| | - Xiaoai Wu
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China,Xiaoai Wu,
| | - Hong Fan
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Hong Fan,
| |
Collapse
|
11
|
Huang Y, Zhang L, Wang M, Li C, Zheng W, Chen H, Liang Y, Wu Z. Optimization of Precursor Synthesis Conditions of (2S,4S)4–[18F]FPArg and Its Application in Glioma Imaging. Pharmaceuticals (Basel) 2022; 15:ph15080946. [PMID: 36015094 PMCID: PMC9416586 DOI: 10.3390/ph15080946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 01/27/2023] Open
Abstract
Although the tracer (2S,4S)4–[18F]FPArg is expected to provide a powerful imaging method for the diagnosis and treatment of clinical tumors, it has not been realized due to the low yield of chemical synthesis and radiolabeling. A simple synthetic method for the radiolabeled precursor of (2S,4S)4–[18F]FPArg in stable yield was obtained by adjusting the sequence of the synthetic steps. Furthermore, the biodistribution experiments confirmed that (2S,4S)4–[18F]FPArg could be cleared out quickly in wild type mouse. Cell uptake experiments and U87MG tumor mouse microPET–CT imaging experiments showed that the tumor had high uptake of (2S,4S)4–[18F]FPArg and the clearance was slow, but (2S,4S)4–[18F]FPArg was rapidly cleared in normal brain tissue. MicroPET–CT imaging of nude mice bearing orthotopic HS683–Luc showed that (2S,4S)4–[18F]FPArg can penetrate blood–brain barrier and image gliomas with a high contrast. Therefore, (2S,4S)4–[18F]FPArg is expected to be further applied in the diagnosis and efficacy evaluation of clinical glioma.
Collapse
Affiliation(s)
- Yong Huang
- Department of Nuclear Medicine, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China; (Y.H.); (C.L.)
| | - Lu Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China; (L.Z.); (W.Z.); (H.C.)
| | - Meng Wang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China;
| | - Chengze Li
- Department of Nuclear Medicine, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China; (Y.H.); (C.L.)
| | - Wei Zheng
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China; (L.Z.); (W.Z.); (H.C.)
| | - Hualong Chen
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China; (L.Z.); (W.Z.); (H.C.)
| | - Ying Liang
- Department of Nuclear Medicine, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China; (Y.H.); (C.L.)
- Correspondence: (Y.L.); (Z.W.)
| | - Zehui Wu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China; (L.Z.); (W.Z.); (H.C.)
- Correspondence: (Y.L.); (Z.W.)
| |
Collapse
|
12
|
Wardak M, Sonni I, Fan AP, Minamimoto R, Jamali M, Hatami N, Zaharchuk G, Fischbein N, Nagpal S, Li G, Koglin N, Berndt M, Bullich S, Stephens AW, Dinkelborg LM, Abel T, Manning HC, Rosenberg J, Chin FT, Sam Gambhir S, Mittra ES. 18F-FSPG PET/CT Imaging of System x C- Transporter Activity in Patients with Primary and Metastatic Brain Tumors. Radiology 2022; 303:620-631. [PMID: 35191738 DOI: 10.1148/radiol.203296] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background The PET tracer (4S)-4-(3-[18F]fluoropropyl)-l-glutamate (18F-FSPG) targets the system xC- cotransporter, which is overexpressed in various tumors. Purpose To assess the role of 18F-FSPG PET/CT in intracranial malignancies. Materials and Methods Twenty-six patients (mean age, 54 years ± 12; 17 men; 48 total lesions) with primary brain tumors (n = 17) or brain metastases (n = 9) were enrolled in this prospective, single-center study (ClinicalTrials.gov identifier: NCT02370563) between November 2014 and March 2016. A 30-minute dynamic brain 18F-FSPG PET/CT scan and a static whole-body (WB) 18F-FSPG PET/CT scan at 60-75 minutes were acquired. Moreover, all participants underwent MRI, and four participants underwent fluorine 18 (18F) fluorodeoxyglucose (FDG) PET imaging. PET parameters and their relative changes were obtained for all lesions. Kinetic modeling was used to estimate the 18F-FSPG tumor rate constants using the dynamic and dynamic plus WB PET data. Imaging parameters were correlated to lesion outcomes, as determined with follow-up MRI and/or pathologic examination. The Mann-Whitney U test or Student t test was used for group mean comparisons. Receiver operating characteristic curve analysis was used for performance comparison of different decision measures. Results 18F-FSPG PET/CT helped identify all 48 brain lesions. The mean tumor-to-background ratio (TBR) on the whole-brain PET images at the WB time point was 26.6 ± 24.9 (range: 2.6-150.3). When 18F-FDG PET was performed, 18F-FSPG permitted visualization of non-18F-FDG-avid lesions or allowed better lesion differentiation from surrounding tissues. In participants with primary brain tumors, the predictive accuracy of the relative changes in influx rate constant Ki and maximum standardized uptake value to discriminate between poor and good lesion outcomes were 89% and 81%, respectively. There were significant differences in the 18F-FSPG uptake curves of lesions with good versus poor outcomes in the primary brain tumor group (P < .05) but not in the brain metastases group. Conclusion PET/CT imaging with (4S)-4-(3-[18F]fluoropropyl)-l-glutamate (18F-FSPG) helped detect primary brain tumors and brain metastases with a high tumor-to-background ratio. Relative changes in 18F-FSPG uptake with multi-time-point PET appear to be helpful in predicting lesion outcomes. Clinical trial registration no. NCT02370563 © RSNA, 2022 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Mirwais Wardak
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Ida Sonni
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Audrey P Fan
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Ryogo Minamimoto
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Mehran Jamali
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Negin Hatami
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Greg Zaharchuk
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Nancy Fischbein
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Seema Nagpal
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Gordon Li
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Norman Koglin
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Mathias Berndt
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Santiago Bullich
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Andrew W Stephens
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Ludger M Dinkelborg
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Ty Abel
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - H Charles Manning
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Jarrett Rosenberg
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Frederick T Chin
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Sanjiv Sam Gambhir
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| | - Erik S Mittra
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (M.W., I.S., A.P.F., R.M., M.J., N.H., G.Z., N.F., J.R., F.T.C., S.S.G., E.S.M.), Department of Neurosurgery (N.F., S.N., G.L.), and Department of Neurology and Neurological Sciences (N.F., S.N., G.L.), Stanford University School of Medicine, Stanford, Calif; Department of Molecular and Medical Pharmacology, UCLA Ahmanson Biological Imaging Center, David Geffen School of Medicine at UCLA, Los Angeles, Calif (I.S.); Department of Biomedical Engineering, Department of Neurology, University of California, Davis, Davis, Calif (A.P.F.); Stanford Bio-X (M.W., G.Z., G.L., F.T.C., S.S.G.) and Departments of Bioengineering (S.S.G.) and Materials Science & Engineering (S.S.G.), Stanford University, Stanford, Calif; Life Molecular Imaging GmbH, Berlin, Germany (N.K., M.B., S.B., A.W.S., L.M.D.); Department of Pathology, Microbiology and Immunology (T.A.) and Department of Radiology and Radiological Sciences, Institute of Imaging Science, Center for Molecular Probes (H.C.M.), Vanderbilt University Medical Center, Nashville, Tenn; and Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Tex (H.C.M.)
| |
Collapse
|
13
|
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
|
14
|
Edwards R, Greenwood HE, McRobbie G, Khan I, Witney TH. Robust and Facile Automated Radiosynthesis of [ 18F]FSPG on the GE FASTlab. Mol Imaging Biol 2021; 23:854-864. [PMID: 34013395 PMCID: PMC8578107 DOI: 10.1007/s11307-021-01609-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/19/2021] [Accepted: 04/18/2021] [Indexed: 01/18/2023]
Abstract
Purpose (S)-4-(3-18F-Fluoropropyl)-ʟ-Glutamic Acid ([18F]FSPG) is a radiolabeled non-natural amino acid that is used for positron emission tomography (PET) imaging of the glutamate/cystine antiporter, system xC-, whose expression is upregulated in many cancer types. To increase the clinical adoption of this radiotracer, reliable and facile automated procedures for [18F]FSPG production are required. Here, we report a cassette-based method to produce [18F]FSPG at high radioactivity concentrations from low amounts of starting activity. Procedures An automated synthesis and purification of [18F]FSPG was developed using the GE FASTlab. Optimization of the reaction conditions and automated manipulations were performed by measuring the isolated radiochemical yield of [18F]FSPG and by assessing radiochemical purity using radio-HPLC. Purification of [18F]FSPG was conducted by trapping and washing of the radiotracer on Oasis MCX SPE cartridges, followed by a reverse elution of [18F]FSPG in phosphate-buffered saline. Subsequently, the [18F]FSPG obtained from the optimized process was used to image an animal model of non-small cell lung cancer. Results The optimized protocol produced [18F]FSPG in 38.4 ± 2.6 % radiochemical yield and >96 % radiochemical purity with a molar activity of 11.1 ± 7.7 GBq/μmol. Small alterations, including the implementation of a reverse elution and an altered Hypercarb cartridge, led to significant improvements in radiotracer concentration from <10 MBq/ml to >100 MBq/ml. The improved radiotracer concentration allowed for the imaging of up to 20 mice, starting with just 1.5 GBq of [18F]Fluoride. Conclusions We have developed a robust and facile method for [18F]FSPG radiosynthesis in high radiotracer concentration, radiochemical yield, and radiochemical purity. This cassette-based method enabled the production of [18F]FSPG at radioactive concentrations sufficient to facilitate large-scale preclinical experiments with a single prep of starting activity. The use of a cassette-based radiosynthesis on an automated synthesis module routinely used for clinical production makes the method amenable to rapid and widespread clinical translation. Supplementary Information The online version contains supplementary material available at 10.1007/s11307-021-01609-w.
Collapse
Affiliation(s)
- Richard Edwards
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital, London, SE1 7EH, UK
| | - Hannah E Greenwood
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital, London, SE1 7EH, UK
| | - Graeme McRobbie
- Pharmaceutical Diagnostics, Life Sciences, GE Healthcare, Pollards Wood, Nightingales Lane, Chalfont St. Giles, Buckinghamshire, HP8 4SP, UK
| | - Imtiaz Khan
- Pharmaceutical Diagnostics, Life Sciences, GE Healthcare, Pollards Wood, Nightingales Lane, Chalfont St. Giles, Buckinghamshire, HP8 4SP, UK
| | - Timothy H Witney
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital, London, SE1 7EH, UK.
| |
Collapse
|
15
|
Park SY, Mosci C, Kumar M, Wardak M, Koglin N, Bullich S, Mueller A, Berndt M, Stephens AW, Chin FT, Gambhir SS, Mittra ES. Initial evaluation of (4S)-4-(3-[ 18F]fluoropropyl)-L-glutamate (FSPG) PET/CT imaging in patients with head and neck cancer, colorectal cancer, or non-Hodgkin lymphoma. EJNMMI Res 2020; 10:100. [PMID: 32857284 PMCID: PMC7455665 DOI: 10.1186/s13550-020-00678-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 07/28/2020] [Indexed: 01/17/2023] Open
Abstract
Purpose (4S)-4-(3-[18F]Fluoropropyl)-l-glutamic acid ([18F]FSPG) measures system xC− transporter activity and shows promise for oncologic imaging. We present data on tumor uptake of this radiopharmaceutical in human subjects with head and neck cancer (HNC), colorectal cancer (CRC), and non-Hodgkin lymphoma (NHL). Methods A total of 15 subjects with HNC (n = 5), CRC (n = 5), or NHL (n = 5) were recruited (mean age 66.2 years, range 44–87 years). 301.4 ± 28.1 MBq (8.1 ± 0.8 mCi) of [18F]FSPG was given intravenously to each subject, and 3 PET/CT scans were obtained 0–2 h post-injection. All subjects also had a positive [18F]FDG PET/CT scan within 1 month prior to the [18F]FSPG PET scan. Semi-quantitative and visual comparisons of the [18F]FSPG and [18F]FDG scans were performed. Results [18F]FSPG showed strong uptake in all but one HNC subject. The lack of surrounding brain uptake facilitated tumor delineation in the HNC patients. [18F]FSPG also showed tumor uptake in all CRC subjects, but variable uptake in the NHL subjects. While the absolute [18F]FDG SUV values were comparable or higher than [18F]FSPG, the tumor-to-background SUV ratios were greater with [18F]FSPG than [18F]FDG. Conclusions [18F]FSPG PET/CT showed promising results across 15 subjects with 3 different cancer types. Concordant visualization was mostly observed between [18F]FSPG and [18F]FDG PET/CT images, with some inter- and intra-individual uptake variability potentially reflecting differences in tumor biology. The tumor-to-background ratios were greater with [18F]FSPG than [18F]FDG in the cancer types evaluated. Future studies based on larger numbers of subjects and those with a wider array of primary and recurrent or metastatic tumors are planned to further evaluate the utility of this novel tracer.
Collapse
Affiliation(s)
- Sonya Y Park
- Department of Radiology, Division of Nuclear Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Camila Mosci
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Meena Kumar
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Mirwais Wardak
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Norman Koglin
- Bayer Pharma AG, Berlin, Germany.,Life Molecular Imaging GmbH, Berlin, Germany
| | | | - Andre Mueller
- Bayer Pharma AG, Berlin, Germany.,Life Molecular Imaging GmbH, Berlin, Germany
| | - Mathias Berndt
- Bayer Pharma AG, Berlin, Germany.,Life Molecular Imaging GmbH, Berlin, Germany
| | - Andrew W Stephens
- Bayer Pharma AG, Berlin, Germany.,Life Molecular Imaging GmbH, Berlin, Germany
| | - Frederick T Chin
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sanjiv S Gambhir
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Bioengineering, Stanford University, Stanford, CA, USA.,Department of Materials Science & Engineering, Stanford University, Stanford, CA, USA.,Bio-X Program, Stanford University, Stanford, CA, USA
| | - Erik S Mittra
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA. .,Department of Diagnostic Radiology, Division of Nuclear Medicine & Molecular Imaging, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Mail Code L340, Portland, OR, 97239, USA.
| |
Collapse
|
16
|
Park SY, Na SJ, Kumar M, Mosci C, Wardak M, Koglin N, Bullich S, Mueller A, Berndt M, Stephens AW, Cho YM, Ahn H, Chae SY, Kim HO, Moon DH, Gambhir SS, Mittra ES. Clinical Evaluation of (4S)-4-(3-[ 18F]Fluoropropyl)-L-glutamate ( 18F-FSPG) for PET/CT Imaging in Patients with Newly Diagnosed and Recurrent Prostate Cancer. Clin Cancer Res 2020; 26:5380-5387. [PMID: 32694158 DOI: 10.1158/1078-0432.ccr-20-0644] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/25/2020] [Accepted: 07/14/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE (4S)-4-(3-[18F]Fluoropropyl)-L-glutamic acid (18F-FSPG) is a radiopharmaceutical for PET imaging of system xC - activity, which can be upregulated in prostate cancer. We present data on the first evaluation of patients with newly diagnosed or recurrent prostate cancer with this radiopharmaceutical. EXPERIMENTAL DESIGN Ten patients with primary and 10 patients with recurrent prostate cancer were enrolled in this prospective multicenter study. After injection of 300 MBq of 18F-FSPG, three whole-body PET/CT scans were obtained. Visual analysis was compared with step-section histopathology when available as well as other imaging studies and clinical outcomes. Metabolic parameters were measured semiquantitatively. Expression levels of xCT and CD44 were evaluated by IHC for patients with available tissue samples. RESULTS 18F-FSPG PET showed high tumor-to-background ratios with a relatively high tumor detection rate on a per-patient (89%) and per-lobe (87%) basis. The sensitivity was slightly higher with imaging at 105 minutes in comparison with 60 minutes. The maximum standardized uptake values (SUVmax) for cancer was significantly higher than both normal (P < 0.005) and benign pathology (P = 0.011), while there was no significant difference between normal and benign pathology (P = 0.120). In the setting of recurrence, agreement with standard imaging was demonstrated in 7 of 9 patients (78%) and 13 of 18 lesions (72%), and revealed true local recurrence in a discordant case. 18F-FSPG accumulation showed moderate correlation with CD44 expression. CONCLUSIONS 18F-FSPG is a promising tumor imaging agent for PET that seems to have favorable biodistribution and high cancer detection rate in patients with prostate cancer. Further studies are warranted to determine the diagnostic value for both initial staging and recurrence, and how it compares with other investigational radiotracers and conventional imaging modalities.
Collapse
Affiliation(s)
- Sonya Youngju Park
- Department of Radiology, College of Medicine, The Catholic University of Korea, Seocho-gu, Seoul, Republic of Korea (South).,Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, California
| | - Sae Jung Na
- Department of Radiology, College of Medicine, The Catholic University of Korea, Seocho-gu, Seoul, Republic of Korea (South).,Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Songpa-gu, Seoul, Republic of Korea (South)
| | - Meena Kumar
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, California
| | - Camila Mosci
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, California
| | - Mirwais Wardak
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, California
| | | | | | | | | | | | - Yong Mee Cho
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Songpa-gu, Seoul, Republic of Korea (South)
| | - Hanjong Ahn
- Department of Urology, Asan Medical Center, University of Ulsan College of Medicine, Songpa-gu, Seoul, Republic of Korea (South)
| | - Sun Young Chae
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Songpa-gu, Seoul, Republic of Korea (South)
| | - Hye Ok Kim
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Songpa-gu, Seoul, Republic of Korea (South).,Department of Nuclear Medicine, Ewha Woman's University College of Medicine, Seodaemun-gu, Seoul, Republic of Korea (South)
| | - Dae Hyuk Moon
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Songpa-gu, Seoul, Republic of Korea (South)
| | - Sanjiv S Gambhir
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, California.,Department of Bioengineering, Department of Materials Science & Engineering, Stanford Bio-X Program, Stanford University, Stanford, California
| | - Erik S Mittra
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, California. .,Department of Diagnostic Radiology, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
17
|
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
|
18
|
Liu S, Wu R, Sun Y, Ploessl K, Zhang Y, Liu Y, Wu Z, Zhu L, Kung HF. Design, synthesis and evaluation of a novel glutamine derivative (2 S,4 R)-2-amino-4-cyano-4-[ 18F]fluorobutanoic acid. NEW J CHEM 2020. [DOI: 10.1039/d0nj00410c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A new glutamine derivative (2S,4R)-2-amino-4-cyano-4-[18F]fluorobutanoic acid (2S,4R)-4-[18F]FCABA ([18F]1) and its labeled precursor can be converted into (2S,4R)-4-[18F]FGln and (2S,4R)4-[18F]FGlu by changing the labeling conditions.
Collapse
Affiliation(s)
- Song Liu
- Beijing Institute of Brain Disorders
- Laboratory of Brain Disorders
- Ministry of Science and Technology
- Collaborative Innovation Center for Brain Disorders
- Capital Medical University
| | - Renbo Wu
- Beijing Institute of Brain Disorders
- Laboratory of Brain Disorders
- Ministry of Science and Technology
- Collaborative Innovation Center for Brain Disorders
- Capital Medical University
| | - Yuli Sun
- Beijing Institute of Brain Disorders
- Laboratory of Brain Disorders
- Ministry of Science and Technology
- Collaborative Innovation Center for Brain Disorders
- Capital Medical University
| | - Karl Ploessl
- Department of Radiology
- University of Pennsylvania
- Philadelphia
- USA
| | - Yan Zhang
- College of Chemistry
- Beijing Normal University
- Beijing
- China
| | - Yajing Liu
- School of Pharmaceutical Science, Capital Medical University
- Beijing 100069
- China
| | - Zehui Wu
- Beijing Institute of Brain Disorders
- Laboratory of Brain Disorders
- Ministry of Science and Technology
- Collaborative Innovation Center for Brain Disorders
- Capital Medical University
| | - Lin Zhu
- College of Chemistry
- Beijing Normal University
- Beijing
- China
| | - Hank F. Kung
- Beijing Institute of Brain Disorders
- Laboratory of Brain Disorders
- Ministry of Science and Technology
- Collaborative Innovation Center for Brain Disorders
- Capital Medical University
| |
Collapse
|
19
|
Wu R, Liu S, Liu Y, Sun Y, Cheng X, Huang Y, Yang Z, Wu Z. Synthesis and biological evaluation of [18F](2S,4S)4-(3-fluoropropyl) arginine as a tumor imaging agent. Eur J Med Chem 2019; 183:111730. [DOI: 10.1016/j.ejmech.2019.111730] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/21/2019] [Accepted: 09/21/2019] [Indexed: 12/31/2022]
|
20
|
Huang Y, Liu S, Wu R, Zhang L, Zhang Y, Hong H, Zhang A, Xiao H, Liu Y, Wu Z, Zhu L, Kung HF. Synthesis and preliminary evaluation of a novel glutamine derivative: (2S,4S)4-[ 18F]FEBGln. Bioorg Med Chem Lett 2019; 29:1047-1050. [PMID: 30871772 DOI: 10.1016/j.bmcl.2019.03.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/04/2019] [Accepted: 03/10/2019] [Indexed: 12/22/2022]
Abstract
We report the preparation of a novel glutamine derivative, (2S,4S)-2,5-diamino-4-(4-(2-fluoroethoxy)benzyl)-5-oxopentanoic acid, (2S, 4S)4-[18F]FEBGln ([18F]4), through efficient organic and radiosyntheses. In vitro assays of [18F]4 using MCF-7 cells showed that it entered cells via multiple amino acid transporter systems including system L and ASC2 transporters but not through the system A transporter. [18F]4 showed promising properties for tumor imaging and may serve as a lead compound for further optimizing and targeting the system L transporter associated with enhanced glutamine metabolism in cancer cells.
Collapse
Affiliation(s)
- Yong Huang
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Song Liu
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Renbo Wu
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Lifang Zhang
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Yan Zhang
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Haiyan Hong
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Aili Zhang
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Hao Xiao
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Yajing Liu
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Zehui Wu
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.
| | - Lin Zhu
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Hank F Kung
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China; Department of Radiology, University of Pennsylvania, Philadelphia 19104, United States.
| |
Collapse
|
21
|
Čolović M, Yang H, Merkens H, Colpo N, Bénard F, Schaffer P. Non-invasive Use of Positron Emission Tomography to Monitor Diethyl maleate and Radiation-Induced Changes in System x C- Activity in Breast Cancer. Mol Imaging Biol 2019; 21:1107-1116. [PMID: 30838549 DOI: 10.1007/s11307-019-01331-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
PURPOSE The system xC- transporter is upregulated in cancer cells in response to oxidative stress (OS). 5-[18F]fluoroaminosuberic acid ([18F]FASu) has been reported as a novel positron emission tomography (PET) imaging agent, targeting system xC-. The goal of this study was to evaluate the utility of [18F]FASu in monitoring cellular response to diethyl maleate (DEM) and radiation-induced OS fluctuations. PROCEDURES [18F]FASu uptake by breast cancer cells was studied in correlation to OS biomarkers: glutathione (GSH) and reactive oxygen species (ROS), as well as transcriptional and translational levels of xCT (the functional subunit of xC-). System xC- inhibitor, sulfasalazine (SSZ), and small interfering RNA (siRNA) knockdown were used as negative controls. Radiotracer uptake was evaluated in three breast cancer models: MDA-MB-231, MCF-7, and ZR-75-1, at two-time points (1 h and 16 h) following OS induction. In vivo [18F]FASu imaging and biodistribution were performed using MDA-MB-231 xenograft-bearing mice at 16 and 24 h post-radiation treatment. RESULTS [18F]FASu uptake was positively correlated to intracellular GSH and SLC7A11 expression levels, and radiotracer uptake was induced both by radiation treatment and by DEM at time points longer than 3 h. In an in vivo setting, there was no statistically significant uptake difference between irradiated and control tumors. CONCLUSION [18F]FASu is a specific system xC- PET radiotracer and as such it can be used to monitor system xC- activity due to OS. As such, [18F]FASu has the potential to be used in therapy response monitoring by PET. Further optimization is required for in vivo application.
Collapse
Affiliation(s)
- Milena Čolović
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC, V6T 2A3, Canada.,British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Hua Yang
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC, V6T 2A3, Canada
| | - Helen Merkens
- British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Nadine Colpo
- British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - François Bénard
- British Columbia Cancer Research Centre, Vancouver, BC, Canada. .,Molecular Oncology, British Columbia Cancer Research Centre, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada. .,Department of Radiology, Faculty of Medicine, University of British Columbia, Vancouver, Canada.
| | - Paul Schaffer
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC, V6T 2A3, Canada. .,Department of Radiology, Faculty of Medicine, University of British Columbia, Vancouver, Canada. .,Department of Chemistry, Faculty of Science, Simon Fraser University, Vancouver, Canada.
| |
Collapse
|
22
|
Prospective comparison of (4S)-4-(3- 18F-fluoropropyl)-L-glutamate versus 18F-fluorodeoxyglucose PET/CT for detecting metastases from pancreatic ductal adenocarcinoma: a proof-of-concept study. Eur J Nucl Med Mol Imaging 2019; 46:810-820. [PMID: 30635754 DOI: 10.1007/s00259-018-4251-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 12/26/2018] [Indexed: 12/22/2022]
Abstract
PURPOSE (4S)-4-(3-18F-Fluoropropyl)-L-glutamate (FSPG) positron emission tomography (PET) reflects system xC- transporter (xCT) expression. FSPG PET has been used to detect brain, lung, breast and liver cancer with only modest success. There is no report on the use of FSPG PET in pancreatic ductal adenocarcinoma (PDAC), presumably because of normal xCT expression in the pancreas. Nonetheless, the tissue-specific expression of xCT in the pancreas suggests that FSPG PET may be ideal for identifying metastasized PDAC. METHODS The performance of FSPG in detecting PDAC metastases was compared with that of 18F-fluorodeoxyglucose (FDG) in small-animal PET studies in seven PDAC tumour-bearing mice and in prospective PET/computed tomography (CT) studies in 23 patients with tissue-confirmed PDAC of stage III or stage IV. All PET/CT results were correlated with the results of histopathology or contrast-enhanced CT (ceCT) performed 3 and 6 months later. RESULTS In the rodent model, FSPG PET consistently found more PDAC metastases earlier than FDG PET. FSPG PET showed a trend for a higher sensitivity, specificity and diagnostic accuracy than FDG PET in detecting PDAC metastases in a patient-based analysis: 95.0%, 100.0% and 95.7%, and 90.0%, 66.7% and 90.0%, respectively. In a lesion-based analysis, FSPG PET identified significantly more PDAC metastases, especially in the liver, than FDG PET (109 vs. 95; P = 0.0001, 95% CI 4.9-14.6). The tumour-to-background ratios for FSPG and FDG uptake on positive scans were similar (FSPG 4.2 ± 4.3, FDG 3.6 ± 3.0; P = 0.44, 95% CI -1.11 to 0.48), despite a lower tumour maximum standardized uptake value in FSPG-avid lesions (FSPG 4.2 + 2.3, FDG 7.7 + 5.7; P = 0.002, 95% CI 0.70-4.10). Because of the lower physiological activity of FSPG in the liver, FSPG PET images of the liver are more easy to interpret than FDG PET images, and therefore the use of FSPG improves the detection of liver metastasis. CONCLUSION FSPG PET is superior to FDG PET in detecting metastasized PDAC, especially in the liver.
Collapse
|
23
|
|
24
|
Zvereva A, Kamp F, Schlattl H, Zankl M, Parodi K. Impact of interpatient variability on organ dose estimates according to MIRD schema: Uncertainty and variance‐based sensitivity analysis. Med Phys 2018; 45:3391-3403. [DOI: 10.1002/mp.12984] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 05/01/2018] [Accepted: 05/08/2018] [Indexed: 11/07/2022] Open
Affiliation(s)
- Alexandra Zvereva
- Institute of Radiation Protection Helmholtz Zentrum München German Research Center for Environmental Health Neuherberg 85764Germany
- Department of Medical Physics Ludwig‐Maximilians‐Universität München (LMU Munich) Garching 85748Germany
| | - Florian Kamp
- Department of Radiation Oncology University Hospital LMU Munich Munich 81377 Germany
| | - Helmut Schlattl
- Institute of Radiation Protection Helmholtz Zentrum München German Research Center for Environmental Health Neuherberg 85764Germany
| | - Maria Zankl
- Institute of Radiation Protection Helmholtz Zentrum München German Research Center for Environmental Health Neuherberg 85764Germany
| | - Katia Parodi
- Department of Medical Physics Ludwig‐Maximilians‐Universität München (LMU Munich) Garching 85748Germany
| |
Collapse
|
25
|
Abstract
PET/CT imaging is frequently used for cancer diagnosis and restaging as metabolically active cells, including cancer, utilize glucose for proliferation. F-FDG is the most commonly utilized radiopharmaceutical in PET/CT imaging. Limitations of F-FDG imaging include intense physiologic uptake in benign tissues such as the brain and myocardium. We present a case of non-small cell lung cancer with myocardial and pericardial metastases obscured by physiologic F-FDG cardiac uptake but detected with the investigational PET radiotracer (4S)-4-(3-F-fluoropropyl)-L-glutamate (F-FSPG), which targets a pathway associated with glutathione biosynthesis. This case demonstrates the added value of F-FSPG PET/CT imaging.
Collapse
|
26
|
Sun A, Liu X, Tang G. Carbon-11 and Fluorine-18 Labeled Amino Acid Tracers for Positron Emission Tomography Imaging of Tumors. Front Chem 2018; 5:124. [PMID: 29379780 PMCID: PMC5775220 DOI: 10.3389/fchem.2017.00124] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 12/12/2017] [Indexed: 12/12/2022] Open
Abstract
Tumor cells have an increased nutritional demand for amino acids (AAs) to satisfy their rapid proliferation. Positron-emitting nuclide labeled AAs are interesting probes and are of great importance for imaging tumors using positron emission tomography (PET). Carbon-11 and fluorine-18 labeled AAs include the [1-11C] AAs, labeling alpha-C- AAs, the branched-chain of AAs and N-substituted carbon-11 labeled AAs. These tracers target protein synthesis or amino acid (AA) transport, and their uptake mechanism mainly involves AA transport. AA PET tracers have been widely used in clinical settings to image brain tumors, neuroendocrine tumors, prostate cancer, breast cancer, non-small cell lung cancer (NSCLC) and hepatocellular carcinoma. This review focuses on the fundamental concepts and the uptake mechanism of AAs, AA PET tracers and their clinical applications.
Collapse
Affiliation(s)
- Aixia Sun
- Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals and Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiang Liu
- Department of Anesthesiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ganghua Tang
- Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals and Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
27
|
Schuster DM, Nanni C, Fanti S. Evaluation of Prostate Cancer with Radiolabeled Amino Acid Analogs. J Nucl Med 2017; 57:61S-66S. [PMID: 27694174 DOI: 10.2967/jnumed.115.170209] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 05/20/2016] [Indexed: 12/14/2022] Open
Abstract
Conventional imaging of prostate cancer has limitations related to the frequently indolent biology of the disease. 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. Radiotracers that are in use or under investigation for targeting salient features of prostate cancer include those directed to glucose, choline, acetate, prostate-specific membrane antigen, bombesin, and amino acids. The tumor imaging features of this last class of radiotracers mirror the upregulation of transmembrane amino acid transport that is necessary in carcinomas because of increased amino acid use for energy requirements and protein synthesis. Natural and synthetic amino acids radiolabeled for PET imaging have been investigated in prostate cancer patients. Early work with naturally occurring amino acid-derived radiotracers, such as l-11C-methionine and l-1-11C-5-hydroxytryptophan, demonstrated promising results, including greater sensitivity than 18F-FDG for intraprostatic and extraprostatic cancer detection. However, limitations with naturally occurring amino acid-derived compounds, including metabolism of the radiotracer itself, led to the development of synthetic amino acid radiotracers, which are not metabolized and therefore more accurately reflect transmembrane amino acid transport. Of the synthetic amino acid-derived PET radiotracers, anti-1-amino-3-18F-fluorocyclobutane-1-carboxylic acid (18F-FACBC or 18F-fluciclovine) has undergone the most promising translation to human use, including the availability of simplified radiosynthesis. Several studies have indicated advantageous biodistribution in the abdomen and pelvis with little renal excretion and bladder activity-characteristics beneficial for prostate cancer imaging. Studies have demonstrated improved lesion detection and diagnostic performance of 18F-fluciclovine in comparison with conventional imaging, especially for recurrent prostate cancer, although issues with nonspecific uptake limit the potential role of 18F-fluciclovine in the diagnosis of primary prostate cancer. Although work is ongoing, recently published intrapatient comparisons of 18F-fluciclovine with 11C-choline reported higher overall diagnostic performance of the former, especially for the detection of disease relapse. This review is aimed at providing a detailed overview of amino acid-derived PET compounds that have been studied for use in prostate cancer imaging.
Collapse
Affiliation(s)
- David M Schuster
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia; and
| | - Cristina Nanni
- Department of Nuclear Medicine, Policlinico S. Orsola, University of Bologna, Bologna, Italy
| | - Stefano Fanti
- Department of Nuclear Medicine, Policlinico S. Orsola, University of Bologna, Bologna, Italy
| |
Collapse
|
28
|
Abstract
Prostate cancer is the most common cancer and the second leading cause of cancer death in men in the United States. Despite high disease prevalence, diagnosis and surveillance of the disease with conventional imaging are limited typically because of indolent biology. Functional imaging with advanced molecular techniques improves the ability to detect disease. Amino acids are building blocks of proteins, and intracellular transport of amino acids is upregulated in prostate cancer. This review provides a detailed overview of the use of F-18 fluciclovine PET in prostate cancer imaging.
Collapse
|
29
|
Abstract
Conventional anatomical imaging with CT and MRI has limitations in the evaluation of prostate cancer. PET is a powerful imaging technique, which can be directed toward molecular targets as diverse as glucose metabolism, density of prostate-specific membrane antigen receptors, and skeletal osteoblastic activity. Although 2-deoxy-2-18F-FDG-PET is the mainstay of molecular imaging, FDG has limitations in typically indolent prostate cancer. Yet, there are many useful and emerging PET tracers beyond FDG, which provide added value. These include radiotracers interrogating prostate cancer via molecular mechanisms related to the biology of choline, acetate, amino acids, bombesin, and dihydrotestosterone, among others. Choline is used for cell membrane synthesis and its metabolism is upregulated in prostate cancer. 11C-choline and 18F-choline are in wide clinical use outside the United States, and they have proven most beneficial for detection of recurrent prostate cancer. 11C-acetate is an indirect biomarker of fatty acid synthesis, which is also upregulated in prostate cancer. Imaging of prostate cancer with 11C-acetate is overall similar to the choline radiotracers yet is not as widely used. Upregulation of amino acid transport in prostate cancer provides the biologic basis for amino acid-based radiotracers. Most recent progress has been made with the nonnatural alicyclic amino acid analogue radiotracer anti-1-amino-3-18F-fluorocyclobutane-1-carboxylic acid (FACBC or fluciclovine) also proven most useful for the detection of recurrent prostate cancer. Other emerging PET radiotracers for prostate cancer include the bombesin group directed to the gastrin-releasing peptide receptor, 16β-18F-fluoro-5α-dihydrotestosterone (FDHT) that binds to the androgen receptor, and those targeting the vasoactive intestinal polypeptide receptor 1 (VPAC-1) and urokinase plasminogen activator receptor (uPAR), which are also overexpressed in prostate cancer.
Collapse
Affiliation(s)
- David M Schuster
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA.
| | - Cristina Nanni
- Department of Nuclear Medicine, Policlinico S. Orsola, University of Bologna, Bologna, Italy
| | - Stefano Fanti
- Department of Nuclear Medicine, Policlinico S. Orsola, University of Bologna, Bologna, Italy
| |
Collapse
|
30
|
Zvereva A, Petoussi-Henss N, Li WB, Schlattl H, Oeh U, Zankl M, Graner FP, Hoeschen C, Nekolla SG, Parodi K, Schwaiger M. Effect of blood activity on dosimetric calculations for radiopharmaceuticals. Phys Med Biol 2016; 61:7688-7703. [DOI: 10.1088/0031-9155/61/21/7688] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
31
|
Pilot Preclinical and Clinical Evaluation of (4S)-4-(3-[18F]Fluoropropyl)-L-Glutamate (18F-FSPG) for PET/CT Imaging of Intracranial Malignancies. PLoS One 2016; 11:e0148628. [PMID: 26890637 PMCID: PMC4758607 DOI: 10.1371/journal.pone.0148628] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 01/19/2016] [Indexed: 01/22/2023] Open
Abstract
Purpose (S)-4-(3-[18F]Fluoropropyl)-L-glutamic acid (18F-FSPG) is a novel radiopharmaceutical for Positron Emission Tomography (PET) imaging. It is a glutamate analogue that can be used to measure xC- transporter activity. This study was performed to assess the feasibility of 18F-FSPG for imaging orthotopic brain tumors in small animals and the translation of this approach in human subjects with intracranial malignancies. Experimental Design For the small animal study, GS9L glioblastoma cells were implanted into brains of Fischer rats and studied with 18F-FSPG, the 18F-labeled glucose derivative 18F-FDG and with the 18F-labeled amino acid derivative 18F-FET. For the human study, five subjects with either primary or metastatic brain cancer were recruited (mean age 50.4 years). After injection of 300 MBq of 18F-FSPG, 3 whole-body PET/Computed Tomography (CT) scans were obtained and safety parameters were measured. The three subjects with brain metastases also had an 18F-FDG PET/CT scan. Quantitative and qualitative comparison of the scans was performed to assess kinetics, biodistribution, and relative efficacy of the tracers. Results In the small animals, the orthotopic brain tumors were visualized well with 18F-FSPG. The high tumor uptake of 18F-FSPG in the GS9L model and the absence of background signal led to good tumor visualization with high contrast (tumor/brain ratio: 32.7). 18F-FDG and 18F-FET showed T/B ratios of 1.7 and 2.8, respectively. In the human pilot study, 18F-FSPG was well tolerated and there was similar distribution in all patients. All malignant lesions were positive with 18F-FSPG except for one low-grade primary brain tumor. In the 18F-FSPG-PET-positive tumors a similar T/B ratio was observed as in the animal model. Conclusions 18F-FSPG is a novel PET radiopharmaceutical that demonstrates good uptake in both small animal and human studies of intracranial malignancies. Future studies on larger numbers of subjects and a wider array of brain tumors are planned. Trial Registration ClinicalTrials.gov NCT01186601
Collapse
|
32
|
Mosci C, Kumar M, Smolarz K, Koglin N, Stephens AW, Schwaiger M, Gambhir SS, Mittra ES. Characterization of Physiologic (18)F FSPG Uptake in Healthy Volunteers. Radiology 2016; 279:898-905. [PMID: 26785040 DOI: 10.1148/radiol.2015142000] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Purpose To evaluate the normal biodistribution and kinetics of (S)-4-(3-[18F]fluoropropyl)-l-glutamic acid ((18)F FSPG) in healthy volunteers and to compare (18)F FSPG mean and maximum standardized uptake values (SUVmean and SUVmax, respectively) with those of (18)F fluorodeoxyglucose (FDG) across a variety of organs. Materials and Methods This protocol was reviewed and approved by all appropriate regulatory authorities. An 8-mCi (±10%) dose of (18)F FSPG was given to five subjects (three women, two men), and seven whole-body positron emission tomography (PET) scans were performed 5, 10, 20, 30, 45, 150, and 240 minutes after injection. Regions of interest were analyzed on the resultant (18)F FSPG images to evaluate the kinetics of this radiotracer. The images obtained 45 minutes after injection were used to measure SUVmean and SUVmax in additional regions of the body. These values were compared with similar values obtained with (18)F FDG PET published previously. Descriptive statistics, including average and standard deviation across the five subjects, were used. (18)F FSPG SUVmean and SUVmax were compared. Results On the (18)F FSPG images obtained 45 minutes after injection, there was only low-grade background activity in the majority of analyzed regions. Prominent activity was seen throughout the pancreas. Clearance of the radiotracer through the kidneys and collection in the bladder also were seen. SUV quantification shows notable differences between (18)F FSPG and (18)F FDG in the pancreas ((18)F FSPG SUVmean, 8.2; (18)F FDG SUVmean, 1.3), stomach ((18)F FSPG SUVmax, 3.6; (18)F FDG SUVmax, 1.6), and brain ((18)F FSPG SUVmean, 0.08; (18)F FDG SUVmean, 7.8). The kinetic data showed rapid clearance of the radiotracer from the blood pool and most organs, except the pancreas. Conclusion (18)F FSPG is a PET radiopharmaceutical characterized by rapid clearance from most healthy tissues, except the pancreas and kidneys. A consistent biodistribution pattern was observed with low background uptake. The physiologic uptake of this new radiotracer throughout the body is described in more detail, which is important for improved interpretative accuracy and understanding potential clinical applications. (©) RSNA, 2016.
Collapse
Affiliation(s)
- Camila Mosci
- From the Molecular Imaging Program, Department of Radiology (C.M., M.K., S.S.G., E.M.), and Bio-X Program, Department of Bioengineering, Department of Materials Science and Engineering (S.S.G.), Stanford University, 300 Pasteur Dr, Stanford, CA 94305-5227; Department of Nuclear Medicine, Technische Universität München, Munich, Germany (K.S., M.S.); and Piramal Imaging, Berlin, Germany (N.K., A.S.)
| | - Meena Kumar
- From the Molecular Imaging Program, Department of Radiology (C.M., M.K., S.S.G., E.M.), and Bio-X Program, Department of Bioengineering, Department of Materials Science and Engineering (S.S.G.), Stanford University, 300 Pasteur Dr, Stanford, CA 94305-5227; Department of Nuclear Medicine, Technische Universität München, Munich, Germany (K.S., M.S.); and Piramal Imaging, Berlin, Germany (N.K., A.S.)
| | - Kamilla Smolarz
- From the Molecular Imaging Program, Department of Radiology (C.M., M.K., S.S.G., E.M.), and Bio-X Program, Department of Bioengineering, Department of Materials Science and Engineering (S.S.G.), Stanford University, 300 Pasteur Dr, Stanford, CA 94305-5227; Department of Nuclear Medicine, Technische Universität München, Munich, Germany (K.S., M.S.); and Piramal Imaging, Berlin, Germany (N.K., A.S.)
| | - Norman Koglin
- From the Molecular Imaging Program, Department of Radiology (C.M., M.K., S.S.G., E.M.), and Bio-X Program, Department of Bioengineering, Department of Materials Science and Engineering (S.S.G.), Stanford University, 300 Pasteur Dr, Stanford, CA 94305-5227; Department of Nuclear Medicine, Technische Universität München, Munich, Germany (K.S., M.S.); and Piramal Imaging, Berlin, Germany (N.K., A.S.)
| | - Andrew W Stephens
- From the Molecular Imaging Program, Department of Radiology (C.M., M.K., S.S.G., E.M.), and Bio-X Program, Department of Bioengineering, Department of Materials Science and Engineering (S.S.G.), Stanford University, 300 Pasteur Dr, Stanford, CA 94305-5227; Department of Nuclear Medicine, Technische Universität München, Munich, Germany (K.S., M.S.); and Piramal Imaging, Berlin, Germany (N.K., A.S.)
| | - Markus Schwaiger
- From the Molecular Imaging Program, Department of Radiology (C.M., M.K., S.S.G., E.M.), and Bio-X Program, Department of Bioengineering, Department of Materials Science and Engineering (S.S.G.), Stanford University, 300 Pasteur Dr, Stanford, CA 94305-5227; Department of Nuclear Medicine, Technische Universität München, Munich, Germany (K.S., M.S.); and Piramal Imaging, Berlin, Germany (N.K., A.S.)
| | - Sanjiv S Gambhir
- From the Molecular Imaging Program, Department of Radiology (C.M., M.K., S.S.G., E.M.), and Bio-X Program, Department of Bioengineering, Department of Materials Science and Engineering (S.S.G.), Stanford University, 300 Pasteur Dr, Stanford, CA 94305-5227; Department of Nuclear Medicine, Technische Universität München, Munich, Germany (K.S., M.S.); and Piramal Imaging, Berlin, Germany (N.K., A.S.)
| | - Erik S Mittra
- From the Molecular Imaging Program, Department of Radiology (C.M., M.K., S.S.G., E.M.), and Bio-X Program, Department of Bioengineering, Department of Materials Science and Engineering (S.S.G.), Stanford University, 300 Pasteur Dr, Stanford, CA 94305-5227; Department of Nuclear Medicine, Technische Universität München, Munich, Germany (K.S., M.S.); and Piramal Imaging, Berlin, Germany (N.K., A.S.)
| |
Collapse
|
33
|
Burkemper JL, Huang C, Li A, Yuan L, Rich K, McConathy J, Lapi SE. Synthesis and Biological Evaluation of (S)-Amino-2-methyl-4-[(76)Br]bromo-3-(E)-butenoic Acid (BrVAIB) for Brain Tumor Imaging. J Med Chem 2015; 58:8542-52. [PMID: 26444035 PMCID: PMC4764504 DOI: 10.1021/acs.jmedchem.5b01035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The novel compound, (S)-amino-2-methyl-4-[(76)Br]bromo-3-(E)-butenoic acid (BrVAIB, [(76)Br]5), was characterized against the known system A tracer, IVAIB ([(123)I]8). [(76)Br]5 was prepared in a 51% ± 19% radiochemical yield with high radiochemical purity (≥98%). The biological properties of [(76)Br]5 were compared with those of [(123)I]8. Results showed that [(76)Br]5 undergoes mixed amino acid transport by system A and system L transport, while [(123)I]8 had less uptake by system L. [(76)Br]5 demonstrated higher uptake than [(123)I]8 in DBT tumors 1 h after injection (3.7 ± 0.4% ID/g vs 1.5 ± 0.3% ID/g) and also showed higher uptake vs [(123)I]8 in normal brain. Small animal PET studies with [(76)Br]5 demonstrated good tumor visualization of intracranial DBTs up to 24 h with clearance from normal tissues. These results indicate that [(76)Br]5 is a promising PET tracer for brain tumor imaging and lead compound for a mixed system A and system L transport substrate.
Collapse
Affiliation(s)
- Jennifer L. Burkemper
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110, United States
| | - Chaofeng Huang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110, United States
| | - Aixiao Li
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110, United States
| | - Liya Yuan
- Department of Neurosurgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110, United States
| | - Keith Rich
- Department of Neurosurgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110, United States
| | - Jonathan McConathy
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110, United States
| | - Suzanne E. Lapi
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110, United States
| |
Collapse
|
34
|
Liu Z, Chen H, Chen K, Shao Y, Kiesewetter DO, Niu G, Chen X. Boramino acid as a marker for amino acid transporters. SCIENCE ADVANCES 2015; 1:e1500694. [PMID: 26601275 PMCID: PMC4643766 DOI: 10.1126/sciadv.1500694] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/08/2015] [Indexed: 06/05/2023]
Abstract
Amino acid transporters (AATs) are a series of integral channels for uphill cellular uptake of nutrients and neurotransmitters. Abnormal expression of AATs is often associated with cancer, addiction, and multiple mental diseases. Although methods to evaluate in vivo expression of AATs would be highly useful, efforts to develop them have been hampered by a lack of appropriate tracers. We describe a new class of AA mimics-boramino acids (BAAs)-that can serve as general imaging probes for AATs. The structure of a BAA is identical to that of the corresponding natural AA, except for an exotic replacement of the carboxylate with -BF3 (-). Cellular studies demonstrate strong AAT-mediated cell uptake, and animal studies show high tumor-specific accumulation, suggesting that BAAs hold great promise for the development of new imaging probes and smart AAT-targeting drugs.
Collapse
Affiliation(s)
- Zhibo Liu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Haojun Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health (NIH), Bethesda, MD 20892, USA
- Department of nuclear medicine, Xiamen Cancer Center, First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Kai Chen
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Yihan Shao
- Laboratory of Computational Biology, National Heart Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Dale O. Kiesewetter
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
35
|
Zanotti-Fregonara P, Lammertsma AA, Innis RB. Suggested pathway to assess radiation safety of ¹⁸F-labeled PET tracers for first-in-human studies. Eur J Nucl Med Mol Imaging 2013; 40:1781-3. [PMID: 23868334 PMCID: PMC3844925 DOI: 10.1007/s00259-013-2512-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 07/02/2013] [Indexed: 11/28/2022]
Affiliation(s)
- Paolo Zanotti-Fregonara
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892-1026, USA
| | - Adriaan A. Lammertsma
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Robert B. Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892-1026, USA
| |
Collapse
|