1
|
Wang H, Qiao X, Ding W, Chen G, Miao Y, Guo R, Zhu X, Cheng Z, Xu J, Li B, Huang Q. Robust and generalizable artificial intelligence for multi-organ segmentation in ultra-low-dose total-body PET imaging: a multi-center and cross-tracer study. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07156-8. [PMID: 39969540 DOI: 10.1007/s00259-025-07156-8] [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: 11/14/2024] [Accepted: 02/12/2025] [Indexed: 02/20/2025]
Abstract
PURPOSE Positron Emission Tomography (PET) is a powerful molecular imaging tool that visualizes radiotracer distribution to reveal physiological processes. Recent advances in total-body PET have enabled low-dose, CT-free imaging; however, accurate organ segmentation using PET-only data remains challenging. This study develops and validates a deep learning model for multi-organ PET segmentation across varied imaging conditions and tracers, addressing critical needs for fully PET-based quantitative analysis. MATERIALS AND METHODS This retrospective study employed a 3D deep learning-based model for automated multi-organ segmentation on PET images acquired under diverse conditions, including low-dose and non-attenuation-corrected scans. Using a dataset of 798 patients from multiple centers with varied tracers, model robustness and generalizability were evaluated via multi-center and cross-tracer tests. Ground-truth labels for 23 organs were generated from CT images, and segmentation accuracy was assessed using the Dice similarity coefficient (DSC). RESULTS In the multi-center dataset from four different institutions, our model achieved average DSC values of 0.834, 0.825, 0.819, and 0.816 across varying dose reduction factors and correction conditions for FDG PET images. In the cross-tracer dataset, the model reached average DSC values of 0.737, 0.573, 0.830, 0.661, and 0.708 for DOTATATE, FAPI, FDG, Grazytracer, and PSMA, respectively. CONCLUSION The proposed model demonstrated effective, fully PET-based multi-organ segmentation across a range of imaging conditions, centers, and tracers, achieving high robustness and generalizability. These findings underscore the model's potential to enhance clinical diagnostic workflows by supporting ultra-low dose PET imaging. CLINICAL TRIAL NUMBER Not applicable. This is a retrospective study based on collected data, which has been approved by the Research Ethics Committee of Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine.
Collapse
Affiliation(s)
- Hanzhong Wang
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Institute for Medical Imaging Technology, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoya Qiao
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Institute for Medical Imaging Technology, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wenxiang Ding
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Gaoyu Chen
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Miao
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Rui Guo
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute for Medical Imaging Technology, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaohua Zhu
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaoping Cheng
- Department of PET/CT, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Jiehua Xu
- Department of Nuclear Medicine, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, China
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Institute for Medical Imaging Technology, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Qiu Huang
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
2
|
Cosmi V, Kvassheim M, Ghosh S, Beekman FJ, Goorden MC. Twisted clustered pinhole collimation for improved high-energy preclinical SPECT/PET. Phys Med Biol 2024; 69:225016. [PMID: 39533753 DOI: 10.1088/1361-6560/ad8c97] [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: 06/18/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Objective.Advanced pinhole collimation geometries optimized for preclinical high-energyɣimaging facilitate applications such asɑandßemitter imaging, simultaneous multi-isotope PET and PET/SPECT, and positron range-free PET. These geometries replace each pinhole with a group of clustered pinholes (CPs) featuring smaller individual pinhole opening angles (POAs), enabling sub-mm resolution imaging up to ∼1 MeV. Further narrowing POAs while retaining field-of-view (FOV) may enhance high-energy imaging but faces geometrical constraints. Here, we detail how the novel twisted CPs (TCPs) address this challenge.Approach.We compared TCP and CP collimator sensitivity at equal system resolution (SR) and SR at matched sensitivity by tuning pinhole diameters for18F (511 keV) and89Zr (909 keV). Additionally, simulated Derenzo phantoms at low activity (LA: 12 MBq ml-1) and high activity (HA: 190 MBq ml-1) levels, along with uniformity images, were compared to assess image resolution and uniformity.Main results.At equal SR, TCP increased average central FOV sensitivity by 15.6% for18F and 29.4% for89Zr compared to CP. Image resolution was comparable, except for89Zr at LA, where TCP resolved 0.80 mm diameter rods compared to 0.90 mm for CP. Image uniformity was equivalent for18F, while for89Zr TCP granted a 10.4% improvement. For collimators with matched sensitivity, TCP improved SR by 6.6% for18F and 17.7% for89Zr while also enhancing image resolution; for18F, rods distinguished were 0.65 mm (CP) and 0.60 mm (TCP) for HA, and 0.70 mm (CP and TCP) for LA. For89Zr, image resolutions were 0.75 mm (CP) and 0.65 mm (TCP) for HA, and 0.90 mm (CP) and 0.80 mm (TCP) for LA. Image uniformity with TCP decreased by 18.3% for18F but improved by 20.1% for89Zr.Significance.This study suggests that the TCP design has potential to improve high-energyɣimaging.
Collapse
Affiliation(s)
- Valerio Cosmi
- Department of Radiation Science and Technology, Delft University of Technology, Delft, The Netherlands
| | - Monika Kvassheim
- Department of Physics and Computational Radiology, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Satyajit Ghosh
- Department of Radiation Science and Technology, Delft University of Technology, Delft, The Netherlands
| | - Freek J Beekman
- Department of Radiation Science and Technology, Delft University of Technology, Delft, The Netherlands
- Free Bee International, Gouda, The Netherlands
| | - Marlies C Goorden
- Department of Radiation Science and Technology, Delft University of Technology, Delft, The Netherlands
| |
Collapse
|
3
|
Yurt F, Özel D, Karagül Ş, Tunçel A, Durkan K, Medine Eİ. 89Zr-Labeled DFO@Durvalumab-HSA nanoparticles: In vitro potential for triple-negative breast cancer. Drug Dev Res 2024; 85:e22266. [PMID: 39363532 DOI: 10.1002/ddr.22266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/13/2024] [Accepted: 09/22/2024] [Indexed: 10/05/2024]
Abstract
This study presents the development and evaluation of a DFO@mAb-NP (DFO@Durvalumab-HSA-DTX nanoparticle) nanoplatform for imaging in triple-negative breast cancer (TNBC). The nanoplatform demonstrated significant changes postconjugation with DFO, evidenced by increased particle size from 178.1 ± 5 nm to 311 ± 26 nm and zeta potential alteration from -31.9 ± 3 mV to -40.5 ± 0.8 mV. Fourier-transform infrared spectroscopy and ultraviolet spectral analyses confirmed successful DFO conjugation, with notable shifts in peak wavelengths. High labeling efficiency was achieved with 89Zr, as indicated by thin layer radio chromatography and high-performance liquid radio chromatography results, with labeling efficiencies of 98 ± 2% for 89Zr-DFO@mAb and 96 ± 3% for 89Zr-DFO@mAb-NP. The nanoplatforms maintained stability over 24 h, showing less than 5% degradation. Lipophilicity assays revealed logP values of 0.5 ± 0.03 for 89Zr-DFO@mAb-NP and 0.98 ± 0.2 for 89Zr-DFO@mAb, indicating a higher lipophilic tendency in the radiolabeled Durvalumab. Cell uptake experiments showed an initial high uptake in MDA-MB-468 cells (45.1 ± 3.2%), which decreased over time, highlighting receptor-specific interactions. These comprehensive findings suggest the promising potential of the DFO@mAb-NP nanoplatform for targeted imaging in TNBC, with implications for improved diagnostic accuracy and treatment strategies.
Collapse
Affiliation(s)
- Fatma Yurt
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova, Izmir, Turkey
- Department of Biomedical Technologies, The Institute of Natural and Applied Sciences, Ege University, Bornova, Izmir, Turkey
| | - Derya Özel
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova, Izmir, Turkey
| | - Şeyma Karagül
- Department of Biomedical Technologies, The Institute of Natural and Applied Sciences, Ege University, Bornova, Izmir, Turkey
| | - Ayça Tunçel
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova, Izmir, Turkey
| | - Kübra Durkan
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova, Izmir, Turkey
| | - Emin İlker Medine
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova, Izmir, Turkey
| |
Collapse
|
4
|
Huang Y, Li Z, Li C, Zang Z, Wang Q, Huang S, Liu Q, Liang Y. Bioorthogonal Diels-Alder Click Chemistry-Based Pretargeted PET Imaging Strategy for Monitoring Programmed Death-Ligand 1 Expression. ACS OMEGA 2024; 9:36969-36981. [PMID: 39246495 PMCID: PMC11375721 DOI: 10.1021/acsomega.4c01063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 07/05/2024] [Accepted: 08/14/2024] [Indexed: 09/10/2024]
Abstract
The development of antibody tracers for positron emission tomography (PET) imaging enables real-time monitoring of tumor expression of programmed cell death ligand 1 (PD-L1) in vivo, aiming to facilitate the selection of immunotherapy responders. However, the slow pharmacokinetics of the antibodies in vivo limits their applications in PET imaging with commonly used radionuclides with short half-lives. In this study, we developed a pretargeted PET imaging strategy based on Diels-Alder (IEDDA) click chemistry to overcome these limitations. Atezolizumab and durvalumab, the most commonly used PD-L1 antibodies in cancer immunotherapy, were selected and compared in the development of the pretargeted PET imaging strategy. Fluorine-18-labeled derivatives of methyl tetrazine ([18F]Tz, [18F]PEG6-Tz, and [18F]PEG12-Tz) were tested in biodistribution and PET imaging of A549-PDL1 xenografts (PD-L1 positive) pretargeted with the trans-cyclooctene (TCO)-functionalized atezolizumab/durvalumab. The biodistribution and imaging results indicated that atezolizumab-TCO/[18F]PEG12-Tz was more suitable for pretargeted PET imaging strategy, and the optimal interval time was 48 h after atezolizumab-TCO administration, where the atezolizumab-TCO/[18F]PEG12-Tz pretargeted approach clearly delineated the A549-PDL1 tumor with a tumor-to-muscle ratio of 5.33, while the ratios are 3.39 and 2.39 for durvalumab/[18F]PEG12-Tz and mock-pretargeting controls, respectively. In conclusion, a pretargeted 18F-immuno-PET imaging technology was successfully established on atezolizumab. The high-contrast PET images of the A549-PDL1 tumor models demonstrate that the pretargeting strategy incorporating short-lived fluorine-18 is viable in identifying tumors with high PD-L1 expression, marking this strategy as a potential candidate for further clinical translation.
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
| | - 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
| | - 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
| | - Zihan Zang
- Shenzhen Middle School, Shenzhen 518024, China
| | - Qiong Wang
- 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
| | - Size 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
| | - Qi Liu
- International Cancer Center, Shenzhen University School of Medicine, Shenzhen University, Shenzhen 518060, 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
| |
Collapse
|
5
|
Tao J, Zeng Z, He C, Meng L, Zhou W, Ren Y, Ma X, Wang Z, Liu J, Li D, Zhang Q, Zhao C, Yang Z, Zhu H. Construction and Preclinical Evaluation of 124I/ 125I-Labeled Antibody Targeting T Cell Immunoglobulin and Mucin Domain-3. Mol Pharm 2024; 21:944-956. [PMID: 38270082 DOI: 10.1021/acs.molpharmaceut.3c01046] [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] [Indexed: 01/26/2024]
Abstract
T cell immunoglobulin and mucin domain-3 (TIM3; HAVCR2) is a transmembrane protein that exerts negative regulatory control over T cell responses. Studies have demonstrated an upregulation of TIM3 expression in tumor-infiltrating lymphocytes (TILs) in cancer patients. In this investigation, a series of monoclonal antibodies targeting TIM3 were produced by hybridoma technology. Among them, C23 exhibited favorable biological properties. To enable specific binding, we developed a 124I/125I-C23 radio-tracer via N-bromosuccinimide (NBS)-mediated labeling of the monoclonal antibody C23. Binding affinity and specificity were assessed using the 293T-TIM3 cell line, which overexpresses TIM3, and the parent 293T cells. Furthermore, biodistribution and in vivo imaging of 124I/125I-C23 were examined in HEK293TIM3 xenograft models and allograft models of 4T1 (mouse breast cancer cells) and CT26 (mouse colon cancer cells). Micro-PET/CT imaging was conducted at intervals of 4, 24, 48, 72, and/or 96 h post intravenous administration of 3.7-7.4 MBq 124I-C23 in the respective model mice. Additionally, immunohistochemistry (IHC) staining of TIM3 expression in dissected tumor organs was performed, along with an assessment of the corresponding expression of Programmed Death 1 (PD1), CD3, and CD8 in the tumors. The C23 monoclonal antibody (mAb) specifically binds to TIM3 protein with a dissociation constant of 23.28 nM. The 124I-C23 and 125I-C23 radio-tracer were successfully prepared with a labeling yield of 83.59 ± 0.35% and 92.35 ± 0.20%, respectively, and over 95.00% radiochemical purity. Stability results indicated that the radiochemical purity of 124I/125I-C23 in phosphate-buffered saline (PBS) and 5% human serum albumin (HSA) was still >80% after 96 h. 125I-C23 uptake in 293T-TIM3 cells was 2.80 ± 0.12%, which was significantly higher than that in 293T cells (1.08 ± 0.08%), and 125I-C23 uptake by 293T-TIM3 cells was significantly blocked at 60 and 120 min in the blocking groups. Pharmacokinetics analysis in vivo revealed an elimination time of 14.62 h and a distribution time of 0.4672 h for 125I-C23. Micro-PET/CT imaging showed that the 124I-C23 probe uptake in the 293T-TIM3 model significantly differed from that of the negative control group and blocking group. In the humanized mouse model, the 124I-C23 probe had obvious specific uptake in the 4T1 and CT26 models and maximum uptake at 24 h in tumor tissues (SUVmax (the maximum standardized uptake value) in 4T1 and CT26 humanized TIM3 murine tumor models: 0.59 ± 0.01 and 0.76 ± 0.02, respectively). Immunohistochemistry of tumor tissues from these mouse models showed comparable TIM3 expression. CD3 and CD8 cells and PD-1 expression were also observed in TIM3-expressing tumor tissues. The TIM3-targeting antibody C23 showed good affinity and specificity. The 124I/125I-C23 probe has obvious targeting specificity for TIM3 in vitro and in vivo. Our results suggest that 124I/125I-C23 is a promising tracer for TIM3 imaging and may have great potential in monitoring immune checkpoint drug efficacy.
Collapse
Affiliation(s)
- Jinping Tao
- School of Medicine, Guizhou University, Guiyang 550025, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ziqing Zeng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Chengxue He
- School of Medicine, Guizhou University, Guiyang 550025, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Lin Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Wenyuan Zhou
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ya'nan Ren
- School of Medicine, Guizhou University, Guiyang 550025, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiaopan Ma
- School of Medicine, Guizhou University, Guiyang 550025, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zilei Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jiayue Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Dapeng Li
- School of Medicine, Guizhou University, Guiyang 550025, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Qian Zhang
- School of Medicine, Guizhou University, Guiyang 550025, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Chuanke Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhi Yang
- School of Medicine, Guizhou University, Guiyang 550025, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Hua Zhu
- School of Medicine, Guizhou University, Guiyang 550025, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
6
|
Ma Y, Gao F. Advances of radiolabeled GRPR ligands for PET/CT imaging of cancers. Cancer Imaging 2024; 24:19. [PMID: 38279185 PMCID: PMC10811881 DOI: 10.1186/s40644-024-00658-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 01/09/2024] [Indexed: 01/28/2024] Open
Abstract
GRPR is a type of seven-transmembrane G-protein coupled receptor that belongs to the bombesin protein receptor family. It is highly expressed in various cancers, including prostate cancer, breast cancer, lung cancer, gastrointestinal cancer, and so on. As a result, molecular imaging studies have been conducted using radiolabeled GRPR ligands for tumor diagnosis, as well as monitoring of recurrence and metastasis. In this paper, we provided a comprehensive overview of relevant literature from the past two decades, with a specific focus on the advancements made in radiolabeled GRPR ligands for imaging prostate cancer and breast cancer.
Collapse
Affiliation(s)
- Yuze Ma
- Research Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Feng Gao
- Research Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
7
|
Lugat A, Bailly C, Chérel M, Rousseau C, Kraeber-Bodéré F, Bodet-Milin C, Bourgeois M. Immuno-PET: Design options and clinical proof-of-concept. Front Med (Lausanne) 2022; 9:1026083. [PMID: 36314010 PMCID: PMC9613928 DOI: 10.3389/fmed.2022.1026083] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/29/2022] [Indexed: 11/23/2022] Open
Abstract
Radioimmunoconjugates have been used for over 30 years in nuclear medicine applications. In the last few years, advances in cancer biology knowledge have led to the identification of new molecular targets specific to certain patient subgroups. The use of these targets in targeted therapies approaches has allowed the developments of specifically tailored therapeutics for patients. As consequence of the PET-imaging progresses, nuclear medicine has developed powerful imaging tools, based on monoclonal antibodies, to in vivo characterization of these tumor biomarkers. This imaging modality known as immuno-positron emission tomography (immuno-PET) is currently in fastest-growing and its medical value lies in its ability to give a non-invasive method to assess the in vivo target expression and distribution and provide key-information on the tumor targeting. Currently, immuno-PET presents promising probes for different nuclear medicine topics as staging/stratification tool, theranostic approaches or predictive/prognostic biomarkers. To develop a radiopharmaceutical drug that can be used in immuno-PET approach, it is necessary to find the best compromise between the isotope choice and the immunologic structure (full monoclonal antibody or derivatives). Through some clinical applications, this paper review aims to discuss the most important aspects of the isotope choice and the usable proteic structure that can be used to meet the clinical needs.
Collapse
Affiliation(s)
- Alexandre Lugat
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France
| | - Clément Bailly
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France
| | - Michel Chérel
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Department of Nuclear Medicine, Institut de Cancérologie de l'Ouest (ICO) – Site Gauducheau, Saint-Herblain, France
| | - Caroline Rousseau
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Department of Nuclear Medicine, Institut de Cancérologie de l'Ouest (ICO) – Site Gauducheau, Saint-Herblain, France
| | - Françoise Kraeber-Bodéré
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France
| | - Caroline Bodet-Milin
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France
| | - Mickaël Bourgeois
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France,ARRONAX Cyclotron, Saint-Herblain, France,*Correspondence: Mickaël Bourgeois
| |
Collapse
|
8
|
Guo R, Xue S, Hu J, Sari H, Mingels C, Zeimpekis K, Prenosil G, Wang Y, Zhang Y, Viscione M, Sznitman R, Rominger A, Li B, Shi K. Using domain knowledge for robust and generalizable deep learning-based CT-free PET attenuation and scatter correction. Nat Commun 2022; 13:5882. [PMID: 36202816 PMCID: PMC9537165 DOI: 10.1038/s41467-022-33562-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Despite the potential of deep learning (DL)-based methods in substituting CT-based PET attenuation and scatter correction for CT-free PET imaging, a critical bottleneck is their limited capability in handling large heterogeneity of tracers and scanners of PET imaging. This study employs a simple way to integrate domain knowledge in DL for CT-free PET imaging. In contrast to conventional direct DL methods, we simplify the complex problem by a domain decomposition so that the learning of anatomy-dependent attenuation correction can be achieved robustly in a low-frequency domain while the original anatomy-independent high-frequency texture can be preserved during the processing. Even with the training from one tracer on one scanner, the effectiveness and robustness of our proposed approach are confirmed in tests of various external imaging tracers on different scanners. The robust, generalizable, and transparent DL development may enhance the potential of clinical translation. Deep learning-based methods have been proposed to substitute CT-based PET attenuation and scatter correction to achieve CT-free PET imaging. Here, the authors present a simple way to integrate domain knowledge in deep learning for CT-free PET imaging.
Collapse
Affiliation(s)
- Rui Guo
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Ruijin Center, Shanghai, China
| | - Song Xue
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Jiaxi Hu
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Hasan Sari
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Advanced Clinical Imaging Technology, Siemens Healthcare AG, Lausanne, Switzerland
| | - Clemens Mingels
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Konstantinos Zeimpekis
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - George Prenosil
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Yue Wang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Ruijin Center, Shanghai, China
| | - Yu Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Ruijin Center, Shanghai, China
| | - Marco Viscione
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Raphael Sznitman
- ARTORG Center, University of Bern, Bern, Switzerland.,Center of Artificial Intelligence in Medicine (CAIM), University of Bern, Bern, Switzerland
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Ruijin Center, Shanghai, China.
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Center of Artificial Intelligence in Medicine (CAIM), University of Bern, Bern, Switzerland.,Computer Aided Medical Procedures and Augmented Reality, Institute of Informatics I16, Technical University of Munich, Munich, Germany
| |
Collapse
|
9
|
Yields of Photo-Proton Reactions on Nuclei of Nickel and Separation of Cobalt Isotopes from Irradiated Targets. Molecules 2022; 27:molecules27051524. [PMID: 35268626 PMCID: PMC8911929 DOI: 10.3390/molecules27051524] [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: 01/25/2022] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 11/16/2022] Open
Abstract
Nowadays, cobalt isotopes 55Co, 57Co, and 58mCo are considered to be promising radionuclides in nuclear medicine, with 55Co receiving the most attention as an isotope for diagnostics by positron emission tomography. One of the current research directions is dedicated to its production using electron accelerators (via photonuclear method). In our work, the yields of nuclear reactions occurring during the irradiation of natNi and 60Ni by bremsstrahlung photons with energy up to 55 MeV were determined. A method of fast and simple cobalt isotopes separation from irradiated targets using extraction chromatography was developed.
Collapse
|
10
|
Kim EE, Youn H, Kang KW. Imaging in Tumor Immunology. Nucl Med Mol Imaging 2021; 55:225-236. [PMID: 34721715 PMCID: PMC8517056 DOI: 10.1007/s13139-021-00706-6] [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: 03/02/2021] [Revised: 05/31/2021] [Accepted: 06/15/2021] [Indexed: 10/20/2022] Open
Abstract
Recent advances in immune modulation have made impressive progress in cancer immunotherapy. Because dynamic nature of the immune response often makes it difficult to evaluate therapeutic outcomes, innovative imaging technologies have been developed to enable non-invasive visualization of immune cells and tumors in their microenvironment. This review summarizes the current tumor immunology and describes new innovative imaging methods with great potential to obtain non-invasive real-time insights into the complex functions of the immune system and into the management of cancer immunotherapy.
Collapse
Affiliation(s)
- Euishin Edmund Kim
- Department of Radiological Sciences, UCI Medical Center, Orange County, CA USA
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Seoul, South Korea
| | - Hyewon Youn
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea
- Laboratory of Molecular Imaging and Therapy, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Keon Wook Kang
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea
- Laboratory of Molecular Imaging and Therapy, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
11
|
George KJH, Borjian S, Cross MC, Hicks JW, Schaffer P, Kovacs MS. Expanding the PET radioisotope universe utilizing solid targets on small medical cyclotrons. RSC Adv 2021; 11:31098-31123. [PMID: 35498914 PMCID: PMC9041346 DOI: 10.1039/d1ra04480j] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/25/2021] [Indexed: 12/17/2022] Open
Abstract
Molecular imaging with medical radioisotopes enables the minimally-invasive monitoring of aberrant biochemical, cellular and tissue-level processes in living subjects. The approach requires the administration of radiotracers composed of radioisotopes attached to bioactive molecules, the pairing of which considers several aspects of the radioisotope in addition to the biological behavior of the targeting molecule to which it is attached. With the advent of modern cellular and biochemical techniques, there has been a virtual explosion in potential disease recognition antigens as well as targeting moieties, which has subsequently opened new applications for a host of emerging radioisotopes with well-matched properties. Additionally, the global radioisotope production landscape has changed rapidly, with reactor-based production and its long-defined, large-scale centralized manufacturing and distribution paradigm shifting to include the manufacture and distribution of many radioisotopes via a worldwide fleet of cyclotrons now in operation. Cyclotron-based radioisotope production has become more prevalent given the commercial availability of instruments, coupled with the introduction of new target hardware, process automation and target manufacturing methods. These advances enable sustained, higher-power irradiation of solid targets that allow hospital-based radiopharmacies to produce a suite of radioisotopes that drive research, clinical trials, and ultimately clinical care. Over the years, several different radioisotopes have been investigated and/or selected for radiolabeling due to favorable decay characteristics (i.e. a suitable half-life, high probability of positron decay, etc.), well-elucidated chemistry, and a feasible production framework. However, longer-lived radioisotopes have surged in popularity given recent regulatory approvals and incorporation of radiopharmaceuticals into patient management within the medical community. This review focuses on the applications, nuclear properties, and production and purification methods for some of the most frequently used/emerging positron-emitting, solid-target-produced radioisotopes that can be manufactured using small-to-medium size cyclotrons (≤24 MeV).
Collapse
Affiliation(s)
- K J H George
- Lawson Health Research Institute 268 Grosvenor Street London ON N6A 4V2 Canada
- Medical Biophysics, Western University 1151 Richmond Street N. London ON N6A 5C1 Canada
| | - S Borjian
- ARTMS 301-4475 Wayburn Drive Burnaby BC V5G 4X4 Canada
| | - M C Cross
- ARTMS 301-4475 Wayburn Drive Burnaby BC V5G 4X4 Canada
| | - J W Hicks
- Lawson Health Research Institute 268 Grosvenor Street London ON N6A 4V2 Canada
- Medical Biophysics, Western University 1151 Richmond Street N. London ON N6A 5C1 Canada
| | - P Schaffer
- Life Sciences, TRIUMF 4004 Wesbrook Mall Vancouver BC V6T 2A3 Canada
- ARTMS 301-4475 Wayburn Drive Burnaby BC V5G 4X4 Canada
- Radiology, University of British Columbia 2775 Laurel St Vancouver BC V5Z 1M9 Canada
- Chemistry, Simon Fraser University 8888 University Dr Burnaby BC V5A 1S6 Canada
| | - M S Kovacs
- Lawson Health Research Institute 268 Grosvenor Street London ON N6A 4V2 Canada
- Medical Biophysics, Western University 1151 Richmond Street N. London ON N6A 5C1 Canada
- Medical Imaging, Western University 1151 Richmond Street N. London ON N6A 5C1 Canada
| |
Collapse
|
12
|
Abstract
Abstract
Purpose
The remarkable amount of preclinical data achieved on 89Zr-PET imaging led to a significant clinical translation, concerning mainly immuno-PET applications. The aim of this systematic review is to provide a complete overview on clinical applications of 89Zr-PET imaging, using a systematic approach to identify and collect published studies performed in humans, sorted by field of application and specific disease subsections.
Methods
A systematic literature search of articles suiting the inclusion criteria was conducted on Pubmed, Scopus, Central, and Web Of Science databases, including papers published from January 1967 to November 2020. Eligible studies had to be performed on humans through PET imaging with 89Zr-labeled compounds. The methodological quality was assessed through the Quality Assessment of Diagnostic accuracy Studies-2 tool.
Results
A total of 821 articles were screened. 74 studies performed on humans were assessed for eligibility with the exclusion of further 18, thus 56 articles were ultimately selected for the qualitative analysis.
Conclusions
89Zr has shown to be a powerful PET-imaging tool, in particular for radiolabeling antibodies in order to study antigen expression, biodistribution, anticancer treatment planning and follow-up. Other than oncologic applications, 89Zr-radiolabeled antibodies have been proposed for use in inflammatory and autoimmune disorders with interesting results. 89Zr-labeled nanoparticles represent groundbreaking radiopharmaceuticals with potential huge fields of application. To evaluate the clinical usefulness of 89Zr PET-imaging in different conditions and in real-world settings, and to widen its use in clinical practice, further translation of preclinical to clinical data is needed.
Collapse
|
13
|
Barrett KE, Houson HA, Lin W, Lapi SE, Engle JW. Production, Purification, and Applications of a Potential Theranostic Pair: Cobalt-55 and Cobalt-58m. Diagnostics (Basel) 2021; 11:diagnostics11071235. [PMID: 34359318 PMCID: PMC8306844 DOI: 10.3390/diagnostics11071235] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 11/16/2022] Open
Abstract
The emerging success of [68Ga/177Lu]Ga/Lu-DOTATATE as a theranostic pair has spurred interest in other isotopes as potential theranostic combinations. Here, we review cobalt-55 and cobalt-58m as a potential theranostic pair. Radionuclidically pure cobalt-55 and cobalt-58m have been produced on small cyclotrons with high molar activity. In vitro, DOTATOC labeled with cobalt has shown greater affinity for SSTR2 than DOTATOC labeled with gallium and yttrium. Similarly, [58mCo]Co-DOTATATE has shown improved cell-killing capabilities as compared to DOTATATE labeled with either indium-111 or lutetium-177. Finally, PET imaging with an isotope such as cobalt-55 allows for image acquisition at much later timepoints than gallium, allowing for an increased degree of biological clearance of non-bound radiotracer. We discuss the accelerator targetry and radiochemistry used to produce cobalt-55,58m, emphasizing the implications of these techniques to downstream radiotracers being developed for imaging and therapy.
Collapse
Affiliation(s)
- Kendall E. Barrett
- Department of Medical Physics, University of Wisconsin, 1111 Highland Avenue, Madison, WI 53711, USA; (K.E.B.); (W.L.)
| | - Hailey A. Houson
- Department of Radiology, University of Alabama at Birmingham, 619 19th Street, Birmingham, AL 35294, USA; (H.A.H.); (S.E.L.)
| | - Wilson Lin
- Department of Medical Physics, University of Wisconsin, 1111 Highland Avenue, Madison, WI 53711, USA; (K.E.B.); (W.L.)
| | - Suzanne E. Lapi
- Department of Radiology, University of Alabama at Birmingham, 619 19th Street, Birmingham, AL 35294, USA; (H.A.H.); (S.E.L.)
| | - Jonathan W. Engle
- Department of Medical Physics, University of Wisconsin, 1111 Highland Avenue, Madison, WI 53711, USA; (K.E.B.); (W.L.)
- Department of Radiology, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792, USA
- Correspondence:
| |
Collapse
|
14
|
Chomet M, van Dongen GAMS, Vugts DJ. State of the Art in Radiolabeling of Antibodies with Common and Uncommon Radiometals for Preclinical and Clinical Immuno-PET. Bioconjug Chem 2021; 32:1315-1330. [PMID: 33974403 PMCID: PMC8299458 DOI: 10.1021/acs.bioconjchem.1c00136] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
Inert
and stable radiolabeling of monoclonal antibodies (mAb),
antibody fragments, or antibody mimetics with radiometals is a prerequisite
for immuno-PET. While radiolabeling is preferably fast, mild, efficient,
and reproducible, especially when applied for human use in a current
Good Manufacturing Practice compliant way, it is crucial that the
obtained radioimmunoconjugate is stable and shows preserved immunoreactivity
and in vivo behavior. Radiometals and chelators have
extensively been evaluated to come to the most ideal radiometal–chelator
pair for each type of antibody derivative. Although PET imaging of
antibodies is a relatively recent tool, applications with 89Zr, 64Cu, and 68Ga have greatly increased in
recent years, especially in the clinical setting, while other less
common radionuclides such as 52Mn, 86Y, 66Ga, and 44Sc, but also 18F as in [18F]AlF are emerging promising candidates for the radiolabeling
of antibodies. This review presents a state of the art overview of
the practical aspects of radiolabeling of antibodies, ranging from
fast kinetic affibodies and nanobodies to slow kinetic intact mAbs.
Herein, we focus on the most common approach which consists of first
modification of the antibody with a chelator, and after eventual storage
of the premodified molecule, radiolabeling as a second step. Other
approaches are possible but have been excluded from this review. The
review includes recent and representative examples from the literature
highlighting which radiometal–chelator–antibody combinations
are the most successful for in vivo application.
Collapse
Affiliation(s)
- Marion Chomet
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam 1081 HV, The Netherlands
| | - Guus A M S van Dongen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam 1081 HV, The Netherlands
| | - Danielle J Vugts
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam 1081 HV, The Netherlands
| |
Collapse
|
15
|
White JM, Escorcia FE, Viola NT. Perspectives on metals-based radioimmunotherapy (RIT): moving forward. Theranostics 2021; 11:6293-6314. [PMID: 33995659 PMCID: PMC8120204 DOI: 10.7150/thno.57177] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/22/2021] [Indexed: 12/18/2022] Open
Abstract
Radioimmunotherapy (RIT) is FDA-approved for the clinical management of liquid malignancies, however, its use for solid malignancies remains a challenge. The putative benefit of RIT lies in selective targeting of antigens expressed on the tumor surface using monoclonal antibodies, to systemically deliver cytotoxic radionuclides. The past several decades yielded dramatic improvements in the quality, quantity, recent commercial availability of alpha-, beta- and Auger Electron-emitting therapeutic radiometals. Investigators have created new or improved existing bifunctional chelators. These bifunctional chelators bind radiometals and can be coupled to antigen-specific antibodies. In this review, we discuss approaches to develop radiometal-based RITs, including the selection of radiometals, chelators and antibody platforms (i.e. full-length, F(ab')2, Fab, minibodies, diabodies, scFv-Fc and nanobodies). We cite examples of the performance of RIT in the clinic, describe challenges to its implementation, and offer insights to address gaps toward translation.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/therapeutic use
- Antigens, Neoplasm/immunology
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/metabolism
- Antineoplastic Agents, Immunological/therapeutic use
- Chelating Agents/administration & dosage
- Chelating Agents/metabolism
- Click Chemistry
- Clinical Trials as Topic
- Dose Fractionation, Radiation
- Drug Delivery Systems
- Forecasting
- Humans
- Immunoglobulin Fab Fragments/administration & dosage
- Immunoglobulin Fab Fragments/therapeutic use
- Lymphoma, Non-Hodgkin/radiotherapy
- Mice
- Molecular Targeted Therapy
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasms, Experimental/diagnostic imaging
- Neoplasms, Experimental/radiotherapy
- Organ Specificity
- Precision Medicine
- Radiation Tolerance
- Radioimmunotherapy/methods
- Radiopharmaceuticals/administration & dosage
- Radiopharmaceuticals/therapeutic use
- Receptor Protein-Tyrosine Kinases/antagonists & inhibitors
- Single-Chain Antibodies/administration & dosage
- Single-Chain Antibodies/therapeutic use
- Single-Domain Antibodies/administration & dosage
- Single-Domain Antibodies/therapeutic use
- Yttrium Radioisotopes/administration & dosage
- Yttrium Radioisotopes/therapeutic use
Collapse
Affiliation(s)
- Jordan M. White
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201
- Department of Oncology, Karmanos Cancer Institute, Detroit, MI 48201
| | - Freddy E. Escorcia
- Molecular Imaging Branch, Radiation Oncology Branch, National Cancer Institute, Bethesda, MD 20814
| | - Nerissa T. Viola
- Department of Oncology, Karmanos Cancer Institute, Detroit, MI 48201
| |
Collapse
|
16
|
Research progress of 18F labeled small molecule positron emission tomography (PET) imaging agents. Eur J Med Chem 2020; 205:112629. [PMID: 32956956 DOI: 10.1016/j.ejmech.2020.112629] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 06/07/2020] [Accepted: 06/28/2020] [Indexed: 01/12/2023]
Abstract
With the development of positron emission tomography (PET) technology, a variety of PET imaging agents labeled with radionuclide 18F have been developed and widely used in the diagnosis and treatment of various clinical diseases in recent years. For example, they have showed a great value of study in the field of tumor detection, tumor treatment and evaluation of tumor therapy in a non-invasive, qualitative and quantitative way. In this review, we highlight the recent development in chemical synthesis, structure and characterization, imaging characterization, and potential applications of these 18F labeled small molecule PET imaging agents for the past five years. The development and application of 18F labeled small molecules will expand our knowledge of the function and distribution of diseases-related molecular targets and shed light on the diagnosis and treatment of various diseases including tumors.
Collapse
|
17
|
Yoon JK, Park BN, Ryu EK, An YS, Lee SJ. Current Perspectives on 89Zr-PET Imaging. Int J Mol Sci 2020; 21:ijms21124309. [PMID: 32560337 PMCID: PMC7352467 DOI: 10.3390/ijms21124309] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/13/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022] Open
Abstract
89Zr is an emerging radionuclide that plays an essential role in immuno-positron emission tomography (PET) imaging. The long half-life of 89Zr (t1/2 = 3.3 days) is favorable for evaluating the in vivo distribution of monoclonal antibodies. Thus, the use of 89Zr is promising for monitoring antibody-based cancer therapies. Immuno-PET combines the sensitivity of PET with the specificity of antibodies. A number of studies have been conducted to investigate the feasibility of 89Zr immuno-PET imaging for predicting the efficacy of radioimmunotherapy and antibody therapies, imaging target expression, detecting target-expressing tumors, and the monitoring of anti-cancer chemotherapies. In this review, we summarize the current status of PET imaging using 89Zr in both preclinical and clinical studies by highlighting the use of immuno-PET for the targets of high clinical relevance. We also present 89Zr-PET applications other than immuno-PET, such as nanoparticle imaging and cell tracking. Finally, we discuss the limitations and the ongoing research being performed to overcome the remaining hurdles.
Collapse
Affiliation(s)
- Joon-Kee Yoon
- Department of Nuclear Medicine & Molecular Imaging, Ajou University School of Medicine, Worldcup-ro 164, Suwon 16499, Korea; (B.-N.P.); (Y.-S.A.); (S.-J.L.)
- Correspondence: ; Tel.: +82-31-219-4303
| | - Bok-Nam Park
- Department of Nuclear Medicine & Molecular Imaging, Ajou University School of Medicine, Worldcup-ro 164, Suwon 16499, Korea; (B.-N.P.); (Y.-S.A.); (S.-J.L.)
| | - Eun-Kyoung Ryu
- Division of Magnetic Resonance, Korea Basic Science Institute, 162, Yeongudanji-ro, Cheongju 28119, Korea;
| | - Young-Sil An
- Department of Nuclear Medicine & Molecular Imaging, Ajou University School of Medicine, Worldcup-ro 164, Suwon 16499, Korea; (B.-N.P.); (Y.-S.A.); (S.-J.L.)
| | - Su-Jin Lee
- Department of Nuclear Medicine & Molecular Imaging, Ajou University School of Medicine, Worldcup-ro 164, Suwon 16499, Korea; (B.-N.P.); (Y.-S.A.); (S.-J.L.)
| |
Collapse
|
18
|
McCarthy CE, White JM, Viola NT, Gibson HM. In vivo Imaging Technologies to Monitor the Immune System. Front Immunol 2020; 11:1067. [PMID: 32582173 PMCID: PMC7280489 DOI: 10.3389/fimmu.2020.01067] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
The past two decades have brought impressive advancements in immune modulation, particularly with the advent of both cancer immunotherapy and biologic therapeutics for inflammatory conditions. However, the dynamic nature of the immune response often complicates the assessment of therapeutic outcomes. Innovative imaging technologies are designed to bridge this gap and allow non-invasive visualization of immune cell presence and/or function in real time. A variety of anatomical and molecular imaging modalities have been applied for this purpose, with each option providing specific advantages and drawbacks. Anatomical methods including magnetic resonance imaging (MRI), computed tomography (CT), and ultrasound provide sharp tissue resolution, which can be further enhanced with contrast agents, including super paramagnetic ions (for MRI) or nanobubbles (for ultrasound). Conjugation of the contrast material to an antibody allows for specific targeting of a cell population or protein of interest. Protein platforms including antibodies, cytokines, and receptor ligands are also popular choices as molecular imaging agents for positron emission tomography (PET), single-photon emission computerized tomography (SPECT), scintigraphy, and optical imaging. These tracers are tagged with either a radioisotope or fluorescent molecule for detection of the target. During the design process for immune-monitoring imaging tracers, it is important to consider any potential downstream physiologic impact. Antibodies may deplete the target cell population, trigger or inhibit receptor signaling, or neutralize the normal function(s) of soluble proteins. Alternatively, the use of cytokines or other ligands as tracers may stimulate their respective signaling pathways, even in low concentrations. As in vivo immune imaging is still in its infancy, this review aims to describe the modalities and immunologic targets that have thus far been explored, with the goal of promoting and guiding the future development and application of novel imaging technologies.
Collapse
Affiliation(s)
- Claire E McCarthy
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Jordan M White
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Nerissa T Viola
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Heather M Gibson
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| |
Collapse
|
19
|
Abstract
Immuno-positron emission tomography (immunoPET) is a paradigm-shifting molecular imaging modality combining the superior targeting specificity of monoclonal antibody (mAb) and the inherent sensitivity of PET technique. A variety of radionuclides and mAbs have been exploited to develop immunoPET probes, which has been driven by the development and optimization of radiochemistry and conjugation strategies. In addition, tumor-targeting vectors with a short circulation time (e.g., Nanobody) or with an enhanced binding affinity (e.g., bispecific antibody) are being used to design novel immunoPET probes. Accordingly, several immunoPET probes, such as 89Zr-Df-pertuzumab and 89Zr-atezolizumab, have been successfully translated for clinical use. By noninvasively and dynamically revealing the expression of heterogeneous tumor antigens, immunoPET imaging is gradually changing the theranostic landscape of several types of malignancies. ImmunoPET is the method of choice for imaging specific tumor markers, immune cells, immune checkpoints, and inflammatory processes. Furthermore, the integration of immunoPET imaging in antibody drug development is of substantial significance because it provides pivotal information regarding antibody targeting abilities and distribution profiles. Herein, we present the latest immunoPET imaging strategies and their preclinical and clinical applications. We also emphasize current conjugation strategies that can be leveraged to develop next-generation immunoPET probes. Lastly, we discuss practical considerations to tune the development and translation of immunoPET imaging strategies.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
| | - Zachary T Rosenkrans
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Quan-Yong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53705, United States
| |
Collapse
|
20
|
Dammes N, Peer D. Monoclonal antibody-based molecular imaging strategies and theranostic opportunities. Theranostics 2020; 10:938-955. [PMID: 31903161 PMCID: PMC6929980 DOI: 10.7150/thno.37443] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 09/26/2019] [Indexed: 01/13/2023] Open
Abstract
Molecular imaging modalities hold great potential as less invasive techniques for diagnosis and management of various diseases. Molecular imaging combines imaging agents with targeting moieties to specifically image diseased sites in the body. Monoclonal antibodies (mAbs) have become increasingly popular as novel therapeutics against a variety of diseases due to their specificity, affinity and serum stability. Because of the same properties, mAbs are also exploited in molecular imaging to target imaging agents such as radionuclides to the cell of interest in vivo. Many studies investigated the use of mAb-targeted imaging for a variety of purposes, for instance to monitor disease progression and to predict response to a specific therapeutic agent. Herein, we highlighted the application of mAb-targeted imaging in three different types of pathologies: autoimmune diseases, oncology and cardiovascular diseases. We also described the potential of molecular imaging strategies in theranostics and precision medicine. Due to the nearly infinite repertoire of mAbs, molecular imaging can change the future of modern medicine by revolutionizing diagnostics and response prediction in practically any disease.
Collapse
Affiliation(s)
- Niels Dammes
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel
- School of Molecular Cell Biology and Biotechnology, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, and Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dan Peer
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel
- School of Molecular Cell Biology and Biotechnology, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, and Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
21
|
Radford LL, Fernandez S, Beacham R, El Sayed R, Farkas R, Benešová M, Müller C, Lapi SE. New 55Co-labeled Albumin-Binding Folate Derivatives as Potential PET Agents for Folate Receptor Imaging. Pharmaceuticals (Basel) 2019; 12:ph12040166. [PMID: 31717279 PMCID: PMC6958329 DOI: 10.3390/ph12040166] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/02/2019] [Accepted: 11/04/2019] [Indexed: 01/23/2023] Open
Abstract
Overexpression of folate receptors (FRs) on different tumor types (e.g., ovarian, lung) make FRs attractive in vivo targets for directed diagnostic/therapeutic agents. Currently, no diagnostic agent suitable for positron emission tomography (PET) has been adopted for clinical FR imaging. In this work, two 55Co-labeled albumin-binding folate derivatives-[55Co]Co-cm10 and [55Co]Co-rf42-with characteristics suitable for PET imaging have been developed and evaluated. High radiochemical yields (≥95%) and in vitro stabilities (≥93%) were achieved for both compounds, and cell assays demonstrated FR-mediated uptake. Both 55Co-labeled folate conjugates demonstrated high tumor uptake of 17% injected activity per gram of tissue (IA/g) at 4 h in biodistribution studies performed in KB tumor-bearing mice. Renal uptake was similar to other albumin-binding folate derivatives, and liver uptake was lower than that of previously reported [64Cu]Cu-rf42. Small animal PET/CT images confirmed the biodistribution results and showed the clear delineation of FR-expressing tumors.
Collapse
Affiliation(s)
- Lauren L. Radford
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (L.L.R.); (S.F.); (R.B.); (R.E.S.)
| | - Solana Fernandez
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (L.L.R.); (S.F.); (R.B.); (R.E.S.)
| | - Rebecca Beacham
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (L.L.R.); (S.F.); (R.B.); (R.E.S.)
| | - Retta El Sayed
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (L.L.R.); (S.F.); (R.B.); (R.E.S.)
| | - Renata Farkas
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland; (R.F.); (M.B.); (C.M.)
| | - Martina Benešová
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland; (R.F.); (M.B.); (C.M.)
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland; (R.F.); (M.B.); (C.M.)
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Suzanne E. Lapi
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (L.L.R.); (S.F.); (R.B.); (R.E.S.)
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35205, USA
- Correspondence:
| |
Collapse
|
22
|
Hervella P, Dam JH, Thisgaard H, Baun C, Olsen BB, Høilund-Carlsen PF, Needham D. Chelation, formulation, encapsulation, retention, and in vivo biodistribution of hydrophobic nanoparticles labelled with 57Co-porphyrin: Oleylamine ensures stable chelation of cobalt in nanoparticles that accumulate in tumors. J Control Release 2018; 291:11-25. [PMID: 30291986 DOI: 10.1016/j.jconrel.2018.09.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 07/02/2018] [Accepted: 09/30/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND MOTIVATION While small molecules can be used in cancer diagnosis there is a need for imageable diagnostic NanoParticles (NPs) that act as surrogates for the therapeutic NPs. Many NPs are composed of hydrophobic materials so the challenge is to formulate hydrophobic imaging agents. To develop individualized medical treatments based on NP, a first step should be the selection of patients who are likely responders to the treatment as judged by imaging tumor accumulation of NPs. This requires NPs with the same size and structure as the subsequent therapeutic NPs but labelled with a long-lived radionuclide. Cobalt isotopes are good candidates for NP labelling since 55Co has half-life of 17.5 h and positron energy of 570 keV while 57Co (t1/2 271.6 d) is an isotope suited for preclinical single photon emission tomography (SPECT) to visualize biodistribution and pharmacokinetics of NPs. We used the hydrophobic octaethyl porphyrin (OEP) to chelate cobalt and to encapsulate it inside hydrophobic liquid NPs (LNPs). We hypothesized that at least two additional hydrophobic axial ligands (oleylamine, OA) must be provided to the OEP-Co complex in order to encapsulate and retain Co inside LNP. RESULTS 1. Cobalt chelation by OEP and OA. The association constant of cobalt to OEP was 2.49 × 105 M-1 and the formation of the hexacoordinate complex OEP-Co-4OA was measured by spectroscopy. 2. NP formulation and characterization: LNPs were prepared by the fast ethanol injection method and were composed of a liquid core (triolein) surrounded by a lipid monolayer (DSPC:Cholesterol:DSPE-PEG2000). The size of the LNPs loaded with the cobalt complex was 40 ± 5 nm, 3. Encapsulation of OEP-Co-OA: The loading capacity of OEP-Co-OA in LNP was 5 mol%. 4. Retention of OEP-57Co-4OA complex in the LNPs: the positive effect of the OA ligands was demonstrated on the stability of the OEP-57Co-4OA complex, providing a half-life for retention in PBS of 170 h (7 days) while in the absence of the axial OA ligands was only 22 h. 5 Biodistribution Study: the in vivo biodistribution of LNP was studied in AR42J pancreatic tumor-bearing mice. The estimated half-life of LNPs in blood was about 7.2 h. Remarkably, the accumulation of LNPs in the tumor was as high as 9.4% ID/g 24 h after injection with a doubling time for tumor accumulation of 3.22 h. The most important result was that the nanoparticles could indeed accumulate in the AR42J tumors up to levels greater than those of other NPs previously measured in the same tumor model, and at about half the values reported for the molecular agent 57Co-DOTATATE. CONCLUSIONS The additional hydrophobic chelator OA was indeed needed to obtain a stable octahedral OEP-Co-4OA. Cobalt was actually well-retained inside LNP in the OEP-Co-4OA complex. The method described in the present work for the core-labelling of LNPs with cobalt is now ready for labeling of NPs with 55Co, or indeed other hexadentate radionuclides of interest for preclinical in vivo PET-imaging and radio-therapeutics.
Collapse
Affiliation(s)
- Pablo Hervella
- Center for Single Particle Science and Engineering (SPSE), Institute for Molecular Medicine, Health Sciences, University Southern Denmark, Campusvej 55, Odense DK-5230, Denmark; Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, Santiago de Compostela 15706, Spain.
| | - Johan Hygum Dam
- Department of Nuclear Medicine, Odense University Hospital, Sdr. Boulevard 29, Odense 5000, Denmark
| | - Helge Thisgaard
- Department of Nuclear Medicine, Odense University Hospital, Sdr. Boulevard 29, Odense 5000, Denmark
| | - Christina Baun
- Department of Nuclear Medicine, Odense University Hospital, Sdr. Boulevard 29, Odense 5000, Denmark
| | - Birgitte Brinkmann Olsen
- Department of Nuclear Medicine, Odense University Hospital, Sdr. Boulevard 29, Odense 5000, Denmark
| | | | - David Needham
- Center for Single Particle Science and Engineering (SPSE), Institute for Molecular Medicine, Health Sciences, University Southern Denmark, Campusvej 55, Odense DK-5230, Denmark; Department of Mechanical Engineering and Material Science, Duke University, Durham, NC 27708,USA; School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK.
| |
Collapse
|
23
|
Carter LM, Poty S, Sharma SK, Lewis JS. Preclinical optimization of antibody-based radiopharmaceuticals for cancer imaging and radionuclide therapy-Model, vector, and radionuclide selection. J Labelled Comp Radiopharm 2018; 61:611-635. [PMID: 29412489 PMCID: PMC6081268 DOI: 10.1002/jlcr.3612] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/19/2017] [Accepted: 01/16/2018] [Indexed: 12/25/2022]
Abstract
Intact antibodies and their truncated counterparts (eg, Fab, scFv fragments) are generally exquisitely specific and selective vectors, enabling recognition of individual cancer-associated molecular phenotypes against a complex and dynamic biomolecular background. Complementary alignment of these advantages with unique properties of radionuclides is a defining paradigm in both radioimmunoimaging and radioimmunotherapy, which remain some of the most adept and promising tools for cancer diagnosis and treatment. This review discusses how translational potency can be maximized through rational selection of antibody-nuclide couples for radioimmunoimaging/therapy in preclinical models.
Collapse
Affiliation(s)
- Lukas M Carter
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sophie Poty
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sai Kiran Sharma
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Departments of Radiology and Pharmacology, Weill Cornell Medical College, New York, New York, USA
- Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
24
|
Aluicio-Sarduy E, Ellison PA, Barnhart TE, Cai W, Nickles RJ, Engle JW. PET radiometals for antibody labeling. J Labelled Comp Radiopharm 2018; 61:636-651. [PMID: 29341227 PMCID: PMC6050152 DOI: 10.1002/jlcr.3607] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 12/29/2017] [Accepted: 01/05/2018] [Indexed: 02/06/2023]
Abstract
Recent advances in molecular characterization of tumors have made possible the emergence of new types of cancer therapies where traditional cytotoxic drugs and nonspecific chemotherapy can be complemented with targeted molecular therapies. One of the main revolutionary treatments is the use of monoclonal antibodies (mAbs) that selectively target the disseminated tumor cells while sparing normal tissues. mAbs and related therapeutics can be efficiently radiolabeled with a wide range of radionuclides to facilitate preclinical and clinical studies. Non-invasive molecular imaging techniques, such as Positron Emission Tomography (PET), using radiolabeled mAbs provide useful information on the whole-body distribution of the biomolecules, which may enable patient stratification, diagnosis, selection of targeted therapies, evaluation of treatment response, and prediction of dose limiting tissue and adverse effects. In addition, when mAbs are labeled with therapeutic radionuclides, the combination of immunological and radiobiological cytotoxicity may result in enhanced treatment efficacy. The pharmacokinetic profile of antibodies demands the use of long half-life isotopes for longitudinal scrutiny of mAb biodistribution and precludes the use of well-stablished short half-life isotopes. Herein, we review the most promising PET radiometals with chemical and physical characteristics that make the appealing for mAb labeling, highlighting those with theranostic radioisotopes.
Collapse
Affiliation(s)
| | - Paul A. Ellison
- University of Wisconsin-Madison, Department of Medical Physics, Madison, Wisconsin, USA
| | - Todd E. Barnhart
- University of Wisconsin-Madison, Department of Medical Physics, Madison, Wisconsin, USA
| | - Weibo Cai
- University of Wisconsin-Madison, Department of Medical Physics, Madison, Wisconsin, USA
- University of Wisconsin-Madison, Department of Radiology, Madison, Wisconsin, USA
- University of Wisconsin-Madison Carbone Cancer Center, Carbon Cancer Center, Madison, Wisconsin, USA
| | - Robert Jerry Nickles
- University of Wisconsin-Madison, Department of Medical Physics, Madison, Wisconsin, USA
| | - Jonathan W. Engle
- University of Wisconsin-Madison, Department of Medical Physics, Madison, Wisconsin, USA
- University of Wisconsin-Madison, Department of Radiology, Madison, Wisconsin, USA
| |
Collapse
|
25
|
Labelling with positron emitters of pnicogens and chalcogens. J Labelled Comp Radiopharm 2017; 61:179-195. [DOI: 10.1002/jlcr.3574] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/07/2017] [Accepted: 09/29/2017] [Indexed: 11/07/2022]
|
26
|
Guérard F, Beyler M, Lee YS, Tripier R, Gestin JF, Brechbiel MW. Investigation of the complexation of natZr(iv) and 89Zr(iv) by hydroxypyridinones for the development of chelators for PET imaging applications. Dalton Trans 2017; 46:4749-4758. [PMID: 28338136 PMCID: PMC5488699 DOI: 10.1039/c6dt04625h] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Three hydroxypyridinone (HOPO) positional isomers - 1,2-HOPO (L1H) and its water soluble analogue (L1'H), 3,2-HOPO (L2H) and 3,4-HOPO (L3H) have been investigated for the complexation of Zr(iv). Potentiometric and UV-Vis spectrometric studies show a higher thermodynamic stability for the formation of Zr(L1')4 in comparison with Zr(L2)4 and Zr(L3)4 as well as a higher kinetic inertness in competition studies with EDTA or Fe3+ at a radiotracer concentration with 89Zr. Besides the low pKa of L1H or L1'H (pKa = 5.01) in comparison with L2H and L3H (pKa = 8.83 and 9.55, respectively), the higher stability of Zr(L1')4 can be attributed in part to the presence of the amide group next to the chelating oxygen that induces intramolecular H-bond and amide/π interactions that were observed by X-ray crystallography and confirmed by quantum chemical calculations. The data presented here indicate that the 1,2-HOPO L1' exhibits the best characteristics for Zr(iv) complexation. However, 3,2-HOPO and 3,4-HOPO patterns, if appropriately tuned, for instance with the addition of an amide group as in the 1,2-HOPO ligand, may also become interesting alternatives for the design of Zr(iv) chelators with improved characteristics for applications in nuclear imaging with 89Zr.
Collapse
Affiliation(s)
- F Guérard
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France. and Radioimmune & Inorganic Chemistry Section, Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - M Beyler
- Université de Bretagne Occidentale, UMR-CNRS 6521 CEMCA, UFR des Sciences et Techniques, Brest, France
| | - Y-S Lee
- Center for Molecular Modeling, Division of Computational Bioscience, Center for Information Technology, National Institutes of Health, Bethesda, Maryland, USA
| | - R Tripier
- Université de Bretagne Occidentale, UMR-CNRS 6521 CEMCA, UFR des Sciences et Techniques, Brest, France
| | - J-F Gestin
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.
| | - M W Brechbiel
- Radioimmune & Inorganic Chemistry Section, Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
27
|
Luo H, England CG, Goel S, Graves SA, Ai F, Liu B, Theuer CP, Wong HC, Nickles RJ, Cai W. ImmunoPET and Near-Infrared Fluorescence Imaging of Pancreatic Cancer with a Dual-Labeled Bispecific Antibody Fragment. Mol Pharm 2017; 14:1646-1655. [PMID: 28292180 DOI: 10.1021/acs.molpharmaceut.6b01123] [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] [Indexed: 11/29/2022]
Abstract
Dual-targeted imaging agents have shown improved targeting efficiencies in comparison to single-targeted entities. The purpose of this study was to quantitatively assess the tumor accumulation of a dual-labeled heterobifunctional imaging agent, targeting two overexpressed biomarkers in pancreatic cancer, using positron emission tomography (PET) and near-infrared fluorescence (NIRF) imaging modalities. A bispecific immunoconjugate (heterodimer) of CD105 and tissue factor (TF) Fab' antibody fragments was developed using click chemistry. The heterodimer was dual-labeled with a radionuclide (64Cu) and fluorescent dye. PET/NIRF imaging and biodistribution studies were performed in four-to-five week old nude athymic mice bearing BxPC-3 (CD105/TF+/+) or PANC-1 (CD105/TF-/-) tumor xenografts. A blocking study was conducted to investigate the specificity of the tracer. Ex vivo tissue staining was performed to compare TF/CD105 expression in tissues with PET tracer uptake to validate in vivo results. PET imaging of 64Cu-NOTA-heterodimer-ZW800 in BxPC-3 tumor xenografts revealed enhanced tumor uptake (21.0 ± 3.4%ID/g; n = 4) compared to the homodimer of TRC-105 (9.6 ± 2.0%ID/g; n = 4; p < 0.01) and ALT-836 (7.6 ± 3.7%ID/g; n = 4; p < 0.01) at 24 h postinjection. Blocking studies revealed that tracer uptake in BxPC-3 tumors could be decreased by 4-fold with TF blocking and 2-fold with CD105 blocking. In the negative model (PANC-1), heterodimer uptake was significantly lower than that found in the BxPC-3 model (3.5 ± 1.1%ID/g; n = 4; p < 0.01). The specificity was confirmed by the successful blocking of CD105 or TF, which demonstrated that the dual targeting with 64Cu-NOTA-heterodimer-ZW800 provided an improvement in overall tumor accumulation. Also, fluorescence imaging validated the PET imaging, allowing for clear delineation of the xenograft tumors. Dual-labeled heterodimeric imaging agents, like 64Cu-NOTA-heterodimer-ZW800, may increase the overall tumor accumulation in comparison to single-targeted homodimers, leading to improved imaging of cancer and other related diseases.
Collapse
Affiliation(s)
- Haiming Luo
- Department of Radiology, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Christopher G England
- Department of Medical Physics, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Shreya Goel
- Materials Science Program, University of Wisconsin-Madison , Madison, Wisconsin 53706, United States
| | - Stephen A Graves
- Department of Medical Physics, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Fanrong Ai
- Department of Radiology, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Bai Liu
- Altor BioScience Corporation , Miramar, Florida 33025, United States
| | - Charles P Theuer
- TRACON Pharmaceuticals Incorporation , San Diego, California 92122, United States
| | - Hing C Wong
- Altor BioScience Corporation , Miramar, Florida 33025, United States
| | - Robert J Nickles
- Department of Medical Physics, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Weibo Cai
- Department of Radiology, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States.,Department of Medical Physics, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States.,Materials Science Program, University of Wisconsin-Madison , Madison, Wisconsin 53706, United States.,University of Wisconsin Carbone Cancer Center , Madison, Wisconsin 53705, United States
| |
Collapse
|
28
|
Bailly C, Cléry PF, Faivre-Chauvet A, Bourgeois M, Guérard F, Haddad F, Barbet J, Chérel M, Kraeber-Bodéré F, Carlier T, Bodet-Milin C. Immuno-PET for Clinical Theranostic Approaches. Int J Mol Sci 2016; 18:ijms18010057. [PMID: 28036044 PMCID: PMC5297692 DOI: 10.3390/ijms18010057] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/16/2016] [Accepted: 12/23/2016] [Indexed: 02/03/2023] Open
Abstract
Recent advances in molecular characterization of tumors have allowed identification of new molecular targets on tumor cells or biomarkers. In medical practice, the identification of these biomarkers slowly but surely becomes a prerequisite before any treatment decision, leading to the concept of personalized medicine. Immuno-positron emission tomography (PET) fits perfectly with this approach. Indeed, monoclonal antibodies (mAbs) labelled with radionuclides represent promising probes for theranostic approaches, offering a non-invasive solution to assess in vivo target expression and distribution. Immuno-PET can potentially provide useful information for patient risk stratification, diagnosis, selection of targeted therapies, evaluation of response to therapy, prediction of adverse effects or for titrating doses for radioimmunotherapy. This paper reviews some aspects and recent developments in labelling methods, biological targets, and clinical data of some novel PET radiopharmaceuticals.
Collapse
Affiliation(s)
- Clément Bailly
- Nantes-Angers Cancer Research Center (CRCNA), University of Nantes, Inserm UMR 892, 8 quai Moncousu, 44007 Nantes, France.
- Department of Nuclear Medicine, CHU de Nantes, 1 place Alexis Ricordeau, 44093 Nantes, France.
| | - Pierre-François Cléry
- Department of Nuclear Medicine, CHU de Nantes, 1 place Alexis Ricordeau, 44093 Nantes, France.
| | - Alain Faivre-Chauvet
- Nantes-Angers Cancer Research Center (CRCNA), University of Nantes, Inserm UMR 892, 8 quai Moncousu, 44007 Nantes, France.
- Department of Nuclear Medicine, CHU de Nantes, 1 place Alexis Ricordeau, 44093 Nantes, France.
| | - Mickael Bourgeois
- Nantes-Angers Cancer Research Center (CRCNA), University of Nantes, Inserm UMR 892, 8 quai Moncousu, 44007 Nantes, France.
- Department of Nuclear Medicine, CHU de Nantes, 1 place Alexis Ricordeau, 44093 Nantes, France.
| | - François Guérard
- Nantes-Angers Cancer Research Center (CRCNA), University of Nantes, Inserm UMR 892, 8 quai Moncousu, 44007 Nantes, France.
| | - Ferid Haddad
- Groupement d'Intérêt Public Arronax, 1, rue Aronnax, CS 10112, 44817 Saint-Herblain, France.
| | - Jacques Barbet
- Groupement d'Intérêt Public Arronax, 1, rue Aronnax, CS 10112, 44817 Saint-Herblain, France.
| | - Michel Chérel
- Nantes-Angers Cancer Research Center (CRCNA), University of Nantes, Inserm UMR 892, 8 quai Moncousu, 44007 Nantes, France.
- Department of Nuclear Medicine, Institut de Cancérologie de l'Ouest (ICO)-René Gauducheau, Boulevard Jacques Monod, 44805 Saint-Herblain, France.
| | - Françoise Kraeber-Bodéré
- Nantes-Angers Cancer Research Center (CRCNA), University of Nantes, Inserm UMR 892, 8 quai Moncousu, 44007 Nantes, France.
- Department of Nuclear Medicine, CHU de Nantes, 1 place Alexis Ricordeau, 44093 Nantes, France.
- Department of Nuclear Medicine, Institut de Cancérologie de l'Ouest (ICO)-René Gauducheau, Boulevard Jacques Monod, 44805 Saint-Herblain, France.
| | - Thomas Carlier
- Nantes-Angers Cancer Research Center (CRCNA), University of Nantes, Inserm UMR 892, 8 quai Moncousu, 44007 Nantes, France.
- Department of Nuclear Medicine, CHU de Nantes, 1 place Alexis Ricordeau, 44093 Nantes, France.
| | - Caroline Bodet-Milin
- Nantes-Angers Cancer Research Center (CRCNA), University of Nantes, Inserm UMR 892, 8 quai Moncousu, 44007 Nantes, France.
- Department of Nuclear Medicine, CHU de Nantes, 1 place Alexis Ricordeau, 44093 Nantes, France.
| |
Collapse
|
29
|
Medved’ko A, Egorova BV, Komarova AA, Rakhimov R, Krut’ko DP, Kalmykov SN, Vatsadze SZ. Copper-Bispidine Complexes: Synthesis and Complex Stability Study. ACS OMEGA 2016; 1:854-867. [PMID: 31457168 PMCID: PMC6640746 DOI: 10.1021/acsomega.6b00237] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 10/20/2016] [Indexed: 05/05/2023]
Abstract
A new series of dicarboxylic derivatives of bispidines have been synthesized to develop novel copper(II) complexes suitable as imaging agents for positron emission tomography. For characterization purposes, copper complexes of bispidines were synthesized in the pure form and in quantitative yields by neutralization of ligands with malachite. The formation of complexes and their stoichiometries were studied by potentiometric titration, cyclic voltammetry, and spectroscopic methods. The stability constants were found to be fairly suitable for copper cation fixation inside dianionic chelate molecules.
Collapse
Affiliation(s)
- Aleksei
V. Medved’ko
- Faculty
of Chemistry and Faculty of Materials Science, Lomonosov
Moscow State University, Leninskie Gory, 1, str. 3, Moscow 119991, Russia
| | - Bayirta V. Egorova
- Faculty
of Chemistry and Faculty of Materials Science, Lomonosov
Moscow State University, Leninskie Gory, 1, str. 3, Moscow 119991, Russia
| | - Alina A. Komarova
- Faculty
of Chemistry and Faculty of Materials Science, Lomonosov
Moscow State University, Leninskie Gory, 1, str. 3, Moscow 119991, Russia
| | - Rustem
D. Rakhimov
- Faculty
of Chemistry and Faculty of Materials Science, Lomonosov
Moscow State University, Leninskie Gory, 1, str. 3, Moscow 119991, Russia
| | - Dmitri P. Krut’ko
- Faculty
of Chemistry and Faculty of Materials Science, Lomonosov
Moscow State University, Leninskie Gory, 1, str. 3, Moscow 119991, Russia
| | - Stepan N. Kalmykov
- Faculty
of Chemistry and Faculty of Materials Science, Lomonosov
Moscow State University, Leninskie Gory, 1, str. 3, Moscow 119991, Russia
| | - Sergey Z. Vatsadze
- Faculty
of Chemistry and Faculty of Materials Science, Lomonosov
Moscow State University, Leninskie Gory, 1, str. 3, Moscow 119991, Russia
- E-mail:
| |
Collapse
|
30
|
Krasikova RN, Aliev RA, Kalmykov SN. The next generation of positron emission tomography radiopharmaceuticals labeled with non-conventional radionuclides. MENDELEEV COMMUNICATIONS 2016. [DOI: 10.1016/j.mencom.2016.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
31
|
|
32
|
Luo H, England CG, Graves SA, Sun H, Liu G, Nickles RJ, Cai W. PET Imaging of VEGFR-2 Expression in Lung Cancer with 64Cu-Labeled Ramucirumab. J Nucl Med 2016; 57:285-90. [PMID: 26541778 PMCID: PMC4738068 DOI: 10.2967/jnumed.115.166462] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 10/21/2015] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED Lung cancer accounts for 17% of cancer-related deaths worldwide, and most patients present with locally advanced or metastatic disease. Novel PET imaging agents for assessing vascular endothelial growth factor receptor-2 (VEGFR-2) expression can be used for detecting VEGFR-2-positive malignancies and subsequent monitoring of therapeutic response to VEGFR-2-targeted therapies. Here, we report the synthesis and characterization of an antibody-based imaging agent for PET imaging of VEGFR-2 expression in vivo. METHODS Ramucirumab (named RamAb), a fully humanized IgG1 monoclonal antibody, was conjugated to 2-S-(4-isothiocyanatobenzyl)-1,4,7-triazacyclononane-1,4,7-triacetic acid (p-SCN-Bn-NOTA) and labeled with (64)Cu. Flow cytometry analysis and microscopy studies were performed to compare the VEGFR-2 binding affinity of RamAb and NOTA-RamAb. PET imaging and biodistribution studies were performed in nude mice bearing HCC4006 and A549 xenograft tumors. Ex vivo histopathology was performed to elucidate the expression patterns of VEGFR-2 in different tissues and organs to validate in vivo results. RESULTS Flow cytometry examination revealed the specific binding capacity of fluorescein isothiocyanate-RamAb to VEGFR-2, and no difference in VEGFR-2 binding affinity was seen between RamAb and NOTA-RamAb. After being labeled with (64)Cu, PET imaging revealed specific and prominent uptake of (64)Cu-NOTA-RamAb in VEGFR-2-positive HCC4006 tumors (9.4 ± 0.5 percentage injected dose per gram at 48 h after injection; n = 4) and significantly lower uptake in VEGFR-2-negative A549 tumors (4.3 ± 0.2 percentage injected dose per gram at 48 h after injection; n = 3). Blocking experiments revealed significantly lower uptake in HCC4006 tumors, along with histology analysis, further confirming the VEGFR-2 specificity of (64)Cu-NOTA-RamAb. CONCLUSION This study provides initial evidence that (64)Cu-NOTA-RamAb can function as a PET imaging agent for visualizing VEGFR-2 expression in vivo, which may also find potential applications in monitoring the treatment response of VEGFR-2-targeted cancer therapy.
Collapse
Affiliation(s)
- Haiming Luo
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Christopher G England
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin; and
| | - Stephen A Graves
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin; and
| | - Haiyan Sun
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Glenn Liu
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - Robert J Nickles
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin; and
| | - Weibo Cai
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin; and University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| |
Collapse
|
33
|
Tailoring medium energy proton beam to induce low energy nuclear reactions in 86SrCl2 for production of PET radioisotope 86Y. Appl Radiat Isot 2015; 101:20-26. [DOI: 10.1016/j.apradiso.2015.02.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 02/23/2015] [Indexed: 11/18/2022]
|
34
|
Makris NE, Boellaard R, van Lingen A, Lammertsma AA, van Dongen GAMS, Verheul HM, Menke CW, Huisman MC. PET/CT-derived whole-body and bone marrow dosimetry of 89Zr-cetuximab. J Nucl Med 2015; 56:249-54. [PMID: 25613538 DOI: 10.2967/jnumed.114.147819] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED PET/CT imaging allows for image-based estimates of organ and red marrow (RM) residence times. The aim of this study was to derive PET/CT-based radiation dosimetry for (89)Zr-cetuximab, with special emphasis on determining RM-absorbed dose. METHODS Seven patients with colorectal cancer received 36.9 ± 0.8 MBq of (89)Zr-cetuximab within 2 h after administration of a therapeutic dose of 500 mg·m(-2) of cetuximab. Whole-body PET/CT scans and blood samples were obtained at 1, 24, 48, 94, and 144 h after injection. RM activity concentrations were calculated from manual delineation of the lumbar vertebrae and blood samples, assuming a fixed RM-to-plasma activity concentration ratio (RMPR) of 0.19. The cumulated activity was calculated as the area under the curve of the organ time-activity data (liver, lungs, kidneys, spleen, and RM), assuming physical decay after the last scan. The residence time for each organ was derived by dividing the cumulated activity with the total injected activity. The residence time in the remainder of the body was calculated as the maximum possible residence time minus the sum of residence time of source organs, assuming no excretion during the time course of the scans. The (self and total) RM- and organ-absorbed doses and effective whole-body radiation dose were obtained using dose conversion factors from OLINDA/EXM 1.1. Several simplified 3-time-point dosimetry approaches were also evaluated. RESULTS The first approach yielded self and total RM doses of 0.17 ± 0.04 and 0.51 ± 0.06 mGy·MBq(-1), respectively. The second approach deviated by -21% in self-dose and -6% in total dose. RMPR increased over time in 5 of 7 patients. The highest (89)Zr-absorbed dose was observed in the liver with 2.60 ± 0.78 mGy·MBq(-1), followed by the kidneys, spleen, and lungs, whereas the effective whole-body dose was 0.61 ± 0.09 mSv·MBq(-1). The simplified 3-time-point (1, 48, and 144 h) dosimetry approach deviated by at most 4% in both organ-absorbed doses and effective dose. CONCLUSION Although the total RM dose estimates obtained with the 2 approaches differed only by at most 6%, the image-based approach is preferred because it accounts for nonconstant RMPR. The number of successive scans can be reduced to 3 without affecting effective dose estimates.
Collapse
Affiliation(s)
- Nikolaos E Makris
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Arthur van Lingen
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Guus A M S van Dongen
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Henk M Verheul
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Catharina W Menke
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Marc C Huisman
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands; and
| |
Collapse
|
35
|
Ocak M, Gillman AG, Bresee J, Zhang L, Vlad AM, Müller C, Schibli R, Edwards WB, Anderson CJ, Gach HM. Folate receptor-targeted multimodality imaging of ovarian cancer in a novel syngeneic mouse model. Mol Pharm 2015; 12:542-53. [PMID: 25536192 PMCID: PMC4319688 DOI: 10.1021/mp500628g] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
A new
transplantable ovarian tumor model is presented using a novel
folate receptor (FR) positive, murine ovarian cancer cell line that
emulates the human disease and induces widespread intraperitoneal
(i.p.) tumors in immunocompetent mice within 4–8 weeks of implantation.
Tumor development was monitored using a new positron emission tomography
(PET) FR-targeting reporter with PET/computerized tomography (PET/CT)
and fluorescence molecular tomography (FMT) using a commercial FR-targeting
reporter. Conventional structural magnetic resonance imaging (MRI)
was also performed. Adult female C57BL/6 mice were injected i.p. with
6 × 106 MKP-L FR+ cells. Imaging was performed weekly
beginning 2 weeks after tumor induction. The albumin-binding, FR-targeting
ligand cm09 was radiolabeled with the positron emitter 68Ga and used to image the tumors with a small animal PET/CT. The FR-reporter
FolateRSense 680 (PerkinElmer) was used for FMT and flow cytometry.
Preclinical MRI (7 T) without FR-targeting was compared with the PET
and FMT molecular imaging. Tumors were visible by all three imaging
modalities. PET/CT had the highest imaging sensitivity at 3–3.5
h postadministration (mean %IA/g mean > 6) and visualized tumors
earlier
than the other two modalities with lower kidney uptake (mean %IA/g
mean < 17) than previously reported FR-targeting agents in late
stage disease. FMT showed relatively low FR-targeted agent in the
bladder and kidneys, but yielded the lowest anatomical image resolution.
MRI produced the highest resolution images, but it was difficult to
distinguish tumors from abdominal organs during early progression
since a FR-targeting MRI reporter was not used. Nevertheless, there
was good correlation of imaging biomarkers between the three modalities.
Tumors in the mouse ovarian cancer model could be detected using FR-targeted
imaging as early as 2 weeks post i.p. injection of tumor cells. An
imaging protocol should combine one or more of the modalities, e.g.,
PET/CT or PET/MRI for optimal tumor detection and delineation from
surrounding tissues.
Collapse
Affiliation(s)
- Meltem Ocak
- Department of Radiology, ‡Cancer Institute, §Department of Obstetrics, Gynecology & Reproductive Sciences, ∥Magee Womens Research Institute, ⊥Department of Pharmacology & Chemical Biology, #Department of Bioengineering, University of Pittsburgh , Pittsburgh, Pennsylvania 15213, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Radchenko V, Busse S, Roesch F. Desferrioxamine as an appropriate chelator for 90Nb: comparison of its complexation properties for M-Df-Octreotide (M = Nb, Fe, Ga, Zr). Nucl Med Biol 2014; 41:721-7. [PMID: 25087170 DOI: 10.1016/j.nucmedbio.2014.06.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 06/10/2014] [Accepted: 06/26/2014] [Indexed: 10/25/2022]
Abstract
The niobium-90 radioisotope ((90)Nb) holds considerable promise for use in immuno-PET, due to its decay parameters (t½ = 14.6h, positron yield=53%, Eß(+)(mean) = 0.35 MeV and Eß(+)(max) = 1.5 MeV). In particular, (90)Nb appears well suited to detect in vivo the pharmacokinetics of large targeting vectors (50-150 kDa). In order to be useful for immuno-PET chelators are required to both stabilize the radionuclide in terms of coordination chemistry and to facilitate the covalent attachment to the targeting vector. Different chelators were evaluated for this purpose in terms of radiolabelling efficiency and stability of the radiolabelled Nb(V) complex and in order to determine the most suitable candidate for conjugation to a biologically relevant targeting vector. For the purpose of studying the complexation properties the niobium radioisotope (95)Nb was used as an analogue of (90)Nb, by virtue of its longer half-life (35 days) and lower cost (reactor-based production). Acyclic and cyclic chelators were investigated, with desferroxamine [Df: (N'-{5-[acetyl(hydroxy)amino]pentyl}-N-[5-({4-[(5-aminopentyl) (hydroxy)amino]-4-oxobutanoyl} amino)pentyl]-N-hydroxysuccinamide)] emerging as the best candidate. Greater than 99% radiolabelling was achieved at room temperature over a wide pH range. The (95)Nb-Df complex is sufficiently stable for immuno-PET (<7% degradation over 7 days in vitro). As a proof-of-principle, a Df conjugate featuring a well-established targeting vector, (D)-Phe(1)-octreotide, was evaluated. The fast labelling kinetics of the unconjugated chelator (Df) were retained for Df-succinyl-(D)Phe(1)-octreotide (Df-OC), with>90% labelling after 1h at room temperature over the pH range 5-7. Stability studies, performed in vitro in serum at physiological temperature (37 °C), revealed that 87 ± 2% of the radiolabelled molecule remained intact after 7 days. Competition studies with relevant metal ions (zirconium((IV)), gallium((III)) and iron((III))) have been performed with Df-OC to gain insight to the relative stability [Nb-Df]-OC complex to transmetallation. At equimolar metal ion concentrations the [Nb-Df]-OC complex showed the greatest overall stability. The favourable radiolabelling characteristics of Df-OC and its stability indicate that Df is a potentially very useful chelator for the development of radiopharmaceuticals for (90)Nb-PET.
Collapse
Affiliation(s)
- Valery Radchenko
- Institute of Nuclear Chemistry, Johannes Gutenberg-University Mainz, Fritz-Strassmann-Weg 2, D-55128 Mainz, Germany.
| | - Stefan Busse
- Institute of Nuclear Chemistry, Johannes Gutenberg-University Mainz, Fritz-Strassmann-Weg 2, D-55128 Mainz, Germany
| | - Frank Roesch
- Institute of Nuclear Chemistry, Johannes Gutenberg-University Mainz, Fritz-Strassmann-Weg 2, D-55128 Mainz, Germany
| |
Collapse
|
37
|
Guérard F, Lee YS, Brechbiel MW. Rational design, synthesis, and evaluation of tetrahydroxamic acid chelators for stable complexation of zirconium(IV). Chemistry 2014; 20:5584-91. [PMID: 24740517 DOI: 10.1002/chem.201304115] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 02/20/2014] [Indexed: 11/06/2022]
Abstract
Metals of interest for biomedical applications often need to be complexed and associated in a stable manner with a targeting agent before use. Whereas the fundamentals of most transition-metal complexation processes have been thoroughly studied, the complexation of Zr(IV) has been somewhat neglected. This metal has received growing attention in recent years, especially in nuclear medicine, with the use of (89) Zr, which a β(+) -emitter with near ideal characteristics for cancer imaging. However, the best chelating agent known for this radionuclide is the trishydroxamate desferrioxamine B (DFB), the Zr(IV) complex of which exhibits suboptimal stability, resulting in the progressive release of (89) Zr in vivo. Based on a recent report demonstrating the higher thermodynamic stability of the tetrahydroxamate complexes of Zr(IV) compared with the trishydroxamate complexes analogues to DFB, we designed a series of tetrahydroxamic acids of varying geometries for improved complexation of this metal. Three macrocycles differing in their cavity size (28 to 36-membered rings) were synthesized by using a ring-closing metathesis strategy, as well as their acyclic analogues. A solution study with (89) Zr showed the complexation to be more effective with increasing cavity size. Evaluation of the kinetic inertness of these new complexes in ethylenediaminetetraacetic acid (EDTA) solution showed significantly improved stabilities of the larger chelates compared with (89) Zr-DFB, whereas the smaller complexes suffered from insufficient stabilities. These results were rationalized by a quantum chemical study. The lower stability of the smaller chelates was attributed to ring strain, whereas the better stability of the larger cyclic complexes was explained by the macrocyclic effect and by the structural rigidity. Overall, these new chelating agents open new perspectives for the safe and efficient use of (89) Zr in nuclear imaging, with the best chelators providing dramatically improved stabilities compared with the reference DFB.
Collapse
Affiliation(s)
- François Guérard
- Radioimmune & Inorganic Chemistry Section, Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892 (USA), Fax: (+1) 301-402-1923.
| | | | | |
Collapse
|
38
|
Zeglis BM, Houghton JL, Evans MJ, Viola-Villegas N, Lewis JS. Underscoring the influence of inorganic chemistry on nuclear imaging with radiometals. Inorg Chem 2014; 53:1880-99. [PMID: 24313747 PMCID: PMC4151561 DOI: 10.1021/ic401607z] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Over the past several decades, radionuclides have matured from largely esoteric and experimental technologies to indispensible components of medical diagnostics. Driving this transition, in part, have been mutually necessary advances in biomedical engineering, nuclear medicine, and cancer biology. Somewhat unsung has been the seminal role of inorganic chemistry in fostering the development of new radiotracers. In this regard, the purpose of this Forum Article is to more visibly highlight the significant contributions of inorganic chemistry to nuclear imaging by detailing the development of five metal-based imaging agents: (64)Cu-ATSM, (68)Ga-DOTATOC, (89)Zr-transferrin, (99m)Tc-sestamibi, and (99m)Tc-colloids. In a concluding section, several unmet needs both in and out of the laboratory will be discussed to stimulate conversation between inorganic chemists and the imaging community.
Collapse
Affiliation(s)
- Brian M. Zeglis
- Department of Radiology and the Program in Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Cancer Center, New York, New York, United States
| | - Jacob L. Houghton
- Department of Radiology and the Program in Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Cancer Center, New York, New York, United States
| | - Michael J. Evans
- Department of Radiology and the Program in Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Cancer Center, New York, New York, United States
| | - Nerissa Viola-Villegas
- Department of Radiology and the Program in Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Cancer Center, New York, New York, United States
| | - Jason S. Lewis
- Department of Radiology and the Program in Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Cancer Center, New York, New York, United States
| |
Collapse
|
39
|
Foerster C, Knight JC, Wuest M, Rowan B, Lapi SE, Amoroso AJ, Edwards PG, Wuest F. Synthesis, complex stability and small animal PET imaging of a novel 64Cu-labelled cryptand molecule. MEDCHEMCOMM 2014. [DOI: 10.1039/c4md00174e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The radiosynthesis and radiopharmacological evaluation including small animal PET imaging of a novel 64Cu-labelled cryptand molecule ([64Cu]CryptTM) possessing a tris-pyridyl/tris-amido set of donor atoms is described.
Collapse
Affiliation(s)
| | | | - Melinda Wuest
- Department of Oncology
- University of Alberta
- Edmonton, Canada
| | - Brendan Rowan
- Cardiff University of Wales – Department of Chemistry
- Cardiff, UK
| | - Suzanne E. Lapi
- Department of Radiology
- Washington University School of Medicine
- St. Louis, USA
| | | | - Peter G. Edwards
- Cardiff University of Wales – Department of Chemistry
- Cardiff, UK
| | - Frank Wuest
- Department of Oncology
- University of Alberta
- Edmonton, Canada
| |
Collapse
|