1
|
Huang Y, Zheng D, Li C, Pi X, Wang S, Li Z, Li Y, Liang Y. Synthesis and preclinical evaluation of an Al 18F radio-fluorinated bivalent PD-L1 nanobody. Eur J Med Chem 2025; 289:117487. [PMID: 40085976 DOI: 10.1016/j.ejmech.2025.117487] [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: 01/22/2025] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Immunotherapy targeting the programmed death 1/programmed death ligand 1 (PD-1/PD-L1) pathway has achieved remarkable clinical success, but there is a shortage of effective approaches for screening suitable patients. Recently developed PD-L1 nanobody probes have limitations, including limited availability of radionuclides, short tumor retention times, and accumulation in non-target organs. To enhance tumor retention and improve tumor-to-normal tissue contrast, we herein report the synthesis and preclinical evaluation of two Al18F-labeled bivalent PD-L1 nanobody probes ([18F]TzTCO-BINb109 and [18F]RESCA-BINb109). Preliminary results indicated that [18F]TzTCO-BINb109 had a greater affinity for PD-L1 and better stability than [18F]RESCA-BINb109. Micro-PET/CT revealed that [18F]TzTCO-BINb109 uptake in A549-PDL1 tumors peaked at 240 min post-injection (3.19 ± 0.49 %ID/g) and demonstrated sustained retention without in vivo defluorination. In contrast, [18F]RESCA-BINb109 exhibited shorter tumor retention (at 60 and 240 min, 2.08 ± 0.22 and 1.37 ± 0.26 %ID/g, respectively) and significant defluorination in vivo. Ex vivo biodistribution studies revealed that the tumor uptake of [18F]TzTCO-BINb109 was consistent with the PET results, with the highest uptake by A549-PDL1 tumor cells (3.43 ± 0.94 %ID/g) compared with H1975 (0.93 ± 0.18 %ID/g) and A549 (0.68 ± 0.12 %ID/g) cells observed at 240 min post-injection. Compared with the previously reported monomeric PD-L1-targeting nanobody probe, [68Ga]NOTA-Nb109, [18F]TzTCO-BINb109 demonstrated enhanced tumor uptake, prolonged retention, and superior tumor-to-normal tissue contrast, contributing to higher imaging quality. These results confirmed that the bivalent PD-L1 nanobody radioligand, [18F]TzTCO-BINb109, was a promising diagnostic probe for PD-L1 detection, efficacy evaluation, and prescription optimization of immune checkpoint inhibitor therapies.
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
| | - Dongye Zheng
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, 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
| | - Xixuan Pi
- Department of Traditional Chinese Medicine, Shenzhen Futian District Maternity & Child Healthcare Hospital, Shenzhen, 518000, China
| | - Senlin 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
| | - 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
| | - Yiluo 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
| | - 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
|
2
|
Martinelli J, Martorana E, Marcotrigiano A, Tei L. Investigations into the N-dealkylation reaction of protected chelating agents. Org Biomol Chem 2025; 23:4090-4099. [PMID: 40171583 DOI: 10.1039/d5ob00114e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
To comply with the specific requirements for the coordination of a certain metal ion, it is often necessary to decrease the substitution degree on the nitrogen atoms of well-known poly(aminocarboxylate) ligands used as chelators for the preparation of diagnostic or therapeutic probes. The procedures used so far to prepare such partially-alkylated compounds involve steps that suffer from product loss or the need to introduce protection/deprotection reactions, consequently lowering the final yield. The application of an N-dealkylation reaction to an exhaustively-substituted precursor could in principle allow to achieve the same result in fewer steps and therefore with higher yields. Dealkylation reactions have been known since the early 1900s, but they have never been exploited for such a purpose. We investigated the applicability of the simple iron-Polonovski N-dealkylation reaction to obtain a library of useful ligands starting from the tert-butyl-protected derivatives of chelators widely used in the biomedical fields such as CDTA, EDTA, NOTA, AAZTA and PCTA. The preparation of partially-alkylated ligands has already been reported in the literature but with several drawbacks and possible improvements. In most of the examples reported, it was found that the reaction occurred in an easy and straightforward way by only using an excess of oxidizing agent that was sufficient to convert the N-oxide into the N-dealkylated product without the need for a reducing agent.
Collapse
Affiliation(s)
- Jonathan Martinelli
- Department of Science and Technological Innovation (DISIT), Università del Piemonte Orientale, Alessandria, Italy.
- Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Enrico Martorana
- Department of Science and Technological Innovation (DISIT), Università del Piemonte Orientale, Alessandria, Italy.
| | - Angelo Marcotrigiano
- Department of Science and Technological Innovation (DISIT), Università del Piemonte Orientale, Alessandria, Italy.
| | - Lorenzo Tei
- Department of Science and Technological Innovation (DISIT), Università del Piemonte Orientale, Alessandria, Italy.
| |
Collapse
|
3
|
Spahn MA, Anbuhl SM, Luyten K, Loy TV, Pronker MF, Cawthorne C, Deroose CM, Schols D, Heukers R, Bormans G, Cleeren F. Indium-111-Labeled Single-Domain Antibody for In Vivo CXCR4 Imaging Using Single-Photon Emission Computed Tomography. Bioconjug Chem 2025; 36:737-747. [PMID: 40067691 DOI: 10.1021/acs.bioconjchem.5c00024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
C-X-C chemokine receptor type 4 (CXCR4) is highly expressed in a range of pathologies, including cancers like multiple myeloma and non-Hodgkin lymphoma, inflammatory diseases such as rheumatoid arthritis, and viral infections like HIV. Currently, the most advanced radiotracer for CXCR4 imaging in clinics is [68Ga]PentixaFor. However, its structure is prone to modifications, complicating the development of a specific CXCR4 fluorine-18-labeled tracer with good pharmacokinetic properties. This study aimed to screen multiple CXCR4-targeting variable domains of heavy-chain-only antibody (VHH or single-domain antibody (sdAb)) constructs to identify the most promising sdAb as a vector molecule for the future development of a CXCR4 fluorine-18 tracer. We have generated five CXCR4-specific sdAb constructs with a cysteine-containing C-terminal tag (C-Direct tag) (VUN400-C-Direct, VUN401-C-Direct, VUN410-C-Direct, VUN411-C-Direct, and VUN415-C-Direct) and one probe (VUN400-C) without. The reduced sdAbs were coupled to maleimide-DOTAGA for 111In-labeling. Their binding affinity against human CXCR4 (hCXCR4) was assessed by using a previously described BRET-based displacement assay. The in vivo profile was assessed using naive mice. Based on the plasma stability (60 min post injection (p.i.)), we selected VUN400-C-Direct and its derivative VUN400-C for further evaluation. These compounds ([111In]In-DOTAGA-VUN400-C-Direct and [111In]In-DOTAGA-VUN400-C) were tested in mice bearing xenografts derived from U87.CD4, U87.CXCR4, and U87.CD4.CXCR4 cells through ex vivo biodistribution studies and SPECT/CT imaging. The six sdAb constructs were labeled with a high radiochemical conversion (75-97%) and purity (>95%). In radioactive binding assays using U87.CD4.CXCR4 cells, [111In]In-DOTAGA-VUN400-C-Direct and [111In]In-DOTAGA-VUN401-C-Direct displayed the highest cellular uptake, achieving 10.4 ± 1.6% and 11.5 ± 1.1%, respectively. In naive mice, [111In]In-DOTAGA-VUN400-C-Direct showed the most favorable biodistribution profile, with low uptake across all organs except the kidneys (Standardized Uptake Value (SUV) > 50, n = 3, 60 min p.i.), but average plasma stability (40.6 ± 9.4%, n = 3, 60 min p.i.). In a xenografted tumor model, [111In]In-DOTAGA-VUN400-C-Direct showed only minor uptake (SUVU87.CXCR4 0.71 ± 0.002, n = 3, 60 min p.i.). [111In]In-DOTAGA-VUN400-C demonstrated nearly identical plasma stability (41.08 ± 5.45%, n = 4) but showed high and specific uptake in the CXCR4-expressing xenografted tumor (SUVU87.CD4.CXCR4 3.75 ± 1.08 vs SUVU87.CD4 = 0.64 ± 0.19, n = 5, 60 min p.i.), which could be blocked by coinjection of AMD3100 (5 mg/kg) (SUVU87.CD4.CXCR4 0.55 ± 0.32 vs SUVU87.CD4 = 0.39 ± 0.07, n = 2, 60 min p.i.). In conclusion, all six sdAbs exhibited high in vitro affinity against hCXCR4. Among these, [111In]In-DOTAGA-VUN400-C showed high CXCR4-specific tumor uptake and favorable pharmacokinetic properties, indicating VUN400-C's potential as a promising vector for future CXCR4 PET imaging applications with fluorine-18.
Collapse
Affiliation(s)
- Muriel Aline Spahn
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven 3000, Belgium
| | - Stephanie Mareike Anbuhl
- QVQ Holding B.V., Yalelaan 1, Utrecht 3584 CL, The Netherlands
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, Amsterdam 1081 HV, The Netherlands
| | - Kaat Luyten
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven 3000, Belgium
| | - Tom Van Loy
- KU Leuven, Department of Microbiology, Immunology and Transplantation Rega Institute for Medical Research,Molecular Structural and Translational Virology Research Group, Leuven B-3000, Belgium
| | - Matti F Pronker
- QVQ Holding B.V., Yalelaan 1, Utrecht 3584 CL, The Netherlands
| | - Christopher Cawthorne
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven 3000, Belgium
| | - Christophe M Deroose
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven 3000, Belgium
| | - Dominique Schols
- KU Leuven, Department of Microbiology, Immunology and Transplantation Rega Institute for Medical Research,Molecular Structural and Translational Virology Research Group, Leuven B-3000, Belgium
| | - Raimond Heukers
- QVQ Holding B.V., Yalelaan 1, Utrecht 3584 CL, The Netherlands
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, Amsterdam 1081 HV, The Netherlands
| | - Guy Bormans
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven 3000, Belgium
| | - Frederik Cleeren
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven 3000, Belgium
| |
Collapse
|
4
|
Zeven K, Dierick H, Sevenois M, Baudhuin H, Navarro L, Van den Block S, Saliën J, Breckpot K, Bridoux J, De Groof TWM, Devoogdt N. Optimizing antibody PET imaging: a comparative preclinical analysis of nanobody and minibody-like PET tracers. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07205-2. [PMID: 40159541 DOI: 10.1007/s00259-025-07205-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025]
Abstract
PURPOSE Positron emission tomography remains the most important medical imaging technique for noninvasive imaging. Advances in radiotracer design, particularly those based on antibody(-formats), have broadened diagnostic applications. While minibodies and nanobodies stand out among antibody-type radiotracers due to their unique characteristics and pharmacokinetics, a direct comparison between these formats remains unexplored. In this study, we generated a so-called minabody, a minibody-like protein consisting of a nanobody fused to a CH3 domain, and compared it with its nanobody counterpart in vitro and in vivo upon labeling with various PET radionuclides. METHODS A previously developed human TIGIT-targeting nanobody was reformatted to a minabody format to allow head-to-head comparison of both formats. The minabody and nanobody formats were compared in vitro for binding, specificity and stability. The minabody and/or nanobody formats were labeled with copper-64, gallium-68 or fluor-18 for non-invasive imaging of human TIGIT-expressing tumors by PET/CT over time. RESULTS The minabody format showed higher binding affinities compared to the nanobody format. Despite lower binding affinity, the nanobody format outperformed the minabody format as PET imaging tracer showing higher specificity and allowing imaging of human TIGIT expression from 1 to 48 h post-injection upon labeling with copper-64. In addition, copper-64 showed superior tumor uptake and contrast for nanobody imaging compared to gallium-68 and fluor-18. CONCLUSION This study describes the first head-to-head comparison between the minibody and nanobody format for PET imaging. Our results underscore the importance of considering both targeting vector format and the radionuclide to achieve accurate and high-quality PET imaging.
Collapse
Affiliation(s)
- Katty Zeven
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, Brussels, Belgium
| | - Herlinde Dierick
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Matthijs Sevenois
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | - Sonja Van den Block
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Jelena Saliën
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, Brussels, Belgium
- Institut de Chimie Moléculaire de l'Université de Bourgogne (ICMUB), UMR CNRS 6302, Université de Bourgogne, Dijon, France
| | - Karine Breckpot
- Translational Oncology Research Center (TORC), Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jessica Bridoux
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, Brussels, Belgium
| | - Timo W M De Groof
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, Brussels, Belgium.
| | - Nick Devoogdt
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
5
|
Kanellopoulos P, Lundmark F, Abouzayed A, Balestri LJI, Håkansson EO, Obeid K, Odell LR, Tolmachev V, Rosenström U, Eriksson J, Orlova A. Synthesis and preclinical evaluation of gastrin releasing peptide receptor antagonist [ 18F]MeTz-PEG 2-RM26 for positron emission tomography. EJNMMI Radiopharm Chem 2025; 10:14. [PMID: 40138074 PMCID: PMC11947346 DOI: 10.1186/s41181-025-00336-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND The gastrin-releasing peptide receptor (GRPR) is overexpressed in the majority of primary prostate cancer lesions, with persistent expression in lymph nodes and bone metastases, making it a legitimate molecular target for diagnostic imaging and staging. This study presents the synthesis and preclinical evaluation of [18F]MeTz-PEG2-RM26, a GRPR antagonist which utilises the Inverse Electron Demand Diels-Alder (IEDDA) reaction for 18F-labelling. This click-chemistry approach allows for site-specific incorporation of fluorine-18 under mild conditions, preserving the peptide's structural integrity and biological activity. Receptor specificity and affinity of [18F]MeTz-PEG2-RM26 were evaluated in vitro using GRPR-expressing PC-3 cells. Furthermore, the biodistribution profile of [18F]MeTz-PEG2-RM26 was assessed in NMRI mice and its tumour-targeting capability was investigated in mice bearing PC-3 xenografts. RESULTS The labelling of TCO-PEG2-RM26 precursor involved three steps: (1) synthesis of an 18F-labelled activated ester on a quaternary methyl ammonium (QMA) cartridge, (2) conjugation of the labelled ester to a tetrazine amine, and (3) attachment to TCO-PEG2-RM26 via an IEDDA click reaction. This production method of [18F]MeTz-PEG2-RM26 afforded a high apparent molar activity of 3.5-4.3 GBq/µmol and radiochemical purity exceeding 98%, with 43-70 MBq activity incorporation, while the entire synthesis was completed within 75 min. Both in vitro and in vivo studies confirmed the specific binding of [18F]MeTz-PEG2-RM26 to GRPR, with a significant reduction in activity uptake observed upon receptor saturation. The radioligand exhibited rapid blood clearance and minimal bone uptake, confirming the stability of the fluorine-carbon bond. However, high hepatic uptake (12-13% IA/g at 1 h post-injection) indicated predominant hepatobiliary excretion. Receptor-mediated uptake was observed in the tumours and pancreatic tissue, although the overall activity uptake in tumours was low, likely due to the rapid hepatobiliary clearance of [18F]MeTz-PEG2-RM26. CONCLUSIONS These findings demonstrate the effectiveness of the IEDDA click reaction for fluorine-18 labelling of GRPR-targeting PET tracers. Future studies should focus on increasing the hydrophilicity of the imaging probe to improve the targeting properties and biodistribution profile of the radioligand.
Collapse
Affiliation(s)
| | - Fanny Lundmark
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Ayman Abouzayed
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | | | | | - Karim Obeid
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Luke R Odell
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ulrika Rosenström
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Jonas Eriksson
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
- PET Centre, Uppsala University Hospital, Uppsala, 751 85, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden.
- Science for Life Laboratory, Uppsala University, Uppsala, 752 37, Sweden.
| |
Collapse
|
6
|
Spreckelmeyer S, Dasilva J, Decristoforo C, Mach RH, Passchier J, Carlucci G, Qhatani MA, Duatti A, Cornelissen BT, Engle J, Denkova A, Hendrikx JJMA, Seimbille Y, Yang X, Jia H, Zhang MR, Yang M, Perk L, Caravan P, Laverman P, Cheng Z, Hoehr C, Sakr T, Zeevaart JR. Highlight selection of radiochemistry and radiopharmacy developments by editorial board. EJNMMI Radiopharm Chem 2025; 10:13. [PMID: 40131608 PMCID: PMC11936851 DOI: 10.1186/s41181-025-00335-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND The Editorial Board of EJNMMI Radiopharmacy and Chemistry releases a biannual highlight commentary to update the readership on trends in the field of radiopharmaceutical development and application of radiopharmaceuticals. MAIN BODY This selection of highlights provides commentary on 24 different topics selected by each co-authoring Editorial Board member addressing a variety of aspects ranging from novel radiochemistry to first-in-human application of novel radiopharmaceuticals. CONCLUSION Trends in radiochemistry and radiopharmacy are highlighted. Hot topics cover the entire scope of EJNMMI Radiopharmacy and Chemistry, demonstrating the progress in the research field in many aspects.
Collapse
Affiliation(s)
- S Spreckelmeyer
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt- Universität zu Berlin, Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - J Dasilva
- University of Montreal, Montreal, Canada
| | | | - R H Mach
- University of Pennsylvania, Philadelphia, USA
| | | | - G Carlucci
- University of California, Los Angeles, CA, USA
| | - M Al Qhatani
- Cyclotron and Radiopharmaceuticals Department, Research & Innovation, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - A Duatti
- University of Ferrara, Ferrara, Italy
| | - B T Cornelissen
- Oxford University, Oxford, UK
- University Medical Center Groningen, Groningen, Netherlands
| | - J Engle
- University of Wisconsin, Madison, WI, USA
| | - A Denkova
- Delft University of Technology, Delft, Netherlands
| | | | | | - X Yang
- Peking University First Hospital, Beijing, China
| | - H Jia
- Beijing Normal University, Beijing, China
| | - M-R Zhang
- National Institutes for Quantum Science and Technology, Chiba, Japan
| | - M Yang
- Jiangsu Institute of Nuclear Medicine, Jiangsu, China
| | - L Perk
- Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| | - P Caravan
- Massuchusetts General Hospital, Harvard University, Boston, USA
| | - P Laverman
- Radboud Medical Center, Nijmegen, Netherlands
| | - Z Cheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - C Hoehr
- TRIUMF, Vancouver, BC, Canada
| | - T Sakr
- Egyptian Atomic Energy Authority, Cairo, Egypt
| | | |
Collapse
|
7
|
Ma X, Hu B, Zhou X, Wang L, Chen H, Xie F, Zhu H, Jia B, Yang Z. Development and First-in-Human evaluation of a Site-Specific [ 18F]-Labeled PD-L1 nanobody PET radiotracer for noninvasive imaging in NSCLC. Bioorg Chem 2025; 156:108222. [PMID: 39889552 DOI: 10.1016/j.bioorg.2025.108222] [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: 12/24/2024] [Revised: 01/13/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Immunohistochemistry (IHC) for PD-L1 detection is limited by its invasiveness and heterogeneity of tumors. To address these challenges, a new PD-L1-targeted nanobody-based immune-PET radiotracer [18F]AlF-APN09 was developed using the site-specific radiolabeling method with the complexing agent (Mal-RESCA) under mild conditions. [18F]AlF-APN09 was prepared at room temperature (pH 4.6-4.8) within 20 min with satisfactory radiochemical yields (45.8 ± 4.48 %, non-decay corrected), high radiochemical purity (>98 %) and moderate apparent molar activity (15-35 GBq/μmol), and remained stable in both PBS and 5 % HSA after 4 h (>90 %). Cell uptake studies indicated variable levels of surface PD-L1 expression in the following order: A549PD-L1 > H1975 > A549. In micro-PET/CT imaging, A549PD-L1 and H1975 tumors were distinctly visualized in a 6.0:1 and 3.2:1 ratios over PD-L1-negative A549 tumors in vivo. Ex vivo biodistribution studies showed tumor uptake values of 6.47 ± 1.06 %ID/g (A549PD-L1) and 2.27 ± 0.19 %ID/g (H1975), significantly higher than 0.90 ± 0.28 %ID/g in A549 tumors. The estimated effective radiation dose in humans was 8.65E-03 mSv/MBq, lower than that of conventional [18F]FDG. First-in-human imaging was conducted on a single resectable non-small cell lung cancer (NSCLC) subject without any adverse reactions. The radiotracer exhibited renal excretion with minimal hepatobiliary clearance. Tumor uptake reached SUVmax 4.20 at 2 h post-injection, demonstrating high contrast and rapid clearance. After PD-1 inhibitor immunotherapy and chemotherapy, the subject showed a therapeutic response and postoperative pathological specimens confirmed a major pathological response (MPR). These results suggest that we have successfully developed a new PD-L1-targeted nanobody PET tracer using the site-specific labeling method with the complexing agent (Mal-RESCA) within 20 min under mild conditions and [18F]AlF-APN09 is a promising noninvasive PET radiotracer for visualizing PD-L1 expression in tumors, offering rapid tumor targeting, excellent signal-to-noise ratios, and favorable clearance properties.
Collapse
Affiliation(s)
- Xiaopan Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), 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; Department of Nuclear Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441138 China
| | - Biao Hu
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191 China
| | - Xin Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), 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
| | - Lei Wang
- Department of Laboratory Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441138 China
| | - Hui Chen
- Department of Nuclear Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441138 China
| | - Fei Xie
- Department of Nuclear Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441138 China
| | - Hua Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), 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.
| | - Bing Jia
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191 China.
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), 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
|
8
|
Zhang Q, Hu Z, Zhao H, Du F, Lv C, Peng T, Zhang Y, Zhang B, Liu J, Wang C. Design, Synthesis, and Biological Evaluation of a Novel [ 18F]AlF-H 3RESCA-FAPI Radiotracer Targeting Fibroblast Activation Protein. Pharmaceuticals (Basel) 2025; 18:277. [PMID: 40006089 PMCID: PMC11859916 DOI: 10.3390/ph18020277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Cancer-associated fibroblasts (CAFs) are key contributors to the tumorigenic process, with fibroblast activation protein (FAP) overexpressed on CAFs in numerous epithelial carcinomas. FAP represents a promising target for tumor imaging and therapy. We aimed to develop a novel [18F]AlF-H3RESCA-FAPI radiotracer with a high labeling yield at room temperature for positron emission tomography (PET) imaging of FAP-expressing tumors. Methods: The H3RESCA-FAPI chelator was synthesized and radiolabeled with [18F]AlF. Its radiotracer binding affinity to FAP was assessed using surface plasmon resonance (SPR). Its in vitro stability, plasma clearance, and biodistribution were evaluated. PET imaging was performed in U87MG tumor-bearing mice, with a blocking study to assess tracer specificity. Results: The [18F]AlF-H3RESCA-FAPI radiotracer demonstrated a high binding affinity to FAP (KD < 10.09 pM) and favorable radiochemical yields (92.4 ± 2.4%) with >95% radiochemical purity. In vitro and in vivo studies showed good stability and rapid clearance from non-target tissues. PET imaging revealed specific tumor uptake, which was significantly reduced by co-injection with unlabeled DOTA-FAPI-04. Conclusions: [18F]AlF-H3RESCA-FAPI is a promising radiotracer for PET imaging of FAP-expressing tumors. Further optimization of its pharmacokinetics could make it a potential candidate for clinical translation.
Collapse
Affiliation(s)
- Qingyu Zhang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; (Q.Z.); (Z.H.); (H.Z.); (F.D.); (C.L.); (T.P.); (Y.Z.)
- MOE Key Lab of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry of Ministry of Education, Shanghai Key Laboratory of Rare Earth Functional Materials, and Shanghai Frontiers Science Centre of Biomimetic Catalysis, Shanghai Normal University, Shanghai 200234, China;
| | - Zhoumi Hu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; (Q.Z.); (Z.H.); (H.Z.); (F.D.); (C.L.); (T.P.); (Y.Z.)
| | - Haitao Zhao
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; (Q.Z.); (Z.H.); (H.Z.); (F.D.); (C.L.); (T.P.); (Y.Z.)
| | - Fuqiang Du
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; (Q.Z.); (Z.H.); (H.Z.); (F.D.); (C.L.); (T.P.); (Y.Z.)
| | - Chun Lv
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; (Q.Z.); (Z.H.); (H.Z.); (F.D.); (C.L.); (T.P.); (Y.Z.)
| | - Tukang Peng
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; (Q.Z.); (Z.H.); (H.Z.); (F.D.); (C.L.); (T.P.); (Y.Z.)
| | - Yukai Zhang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; (Q.Z.); (Z.H.); (H.Z.); (F.D.); (C.L.); (T.P.); (Y.Z.)
- MOE Key Lab of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry of Ministry of Education, Shanghai Key Laboratory of Rare Earth Functional Materials, and Shanghai Frontiers Science Centre of Biomimetic Catalysis, Shanghai Normal University, Shanghai 200234, China;
| | - Bowu Zhang
- MOE Key Lab of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry of Ministry of Education, Shanghai Key Laboratory of Rare Earth Functional Materials, and Shanghai Frontiers Science Centre of Biomimetic Catalysis, Shanghai Normal University, Shanghai 200234, China;
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; (Q.Z.); (Z.H.); (H.Z.); (F.D.); (C.L.); (T.P.); (Y.Z.)
| | - Cheng Wang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; (Q.Z.); (Z.H.); (H.Z.); (F.D.); (C.L.); (T.P.); (Y.Z.)
| |
Collapse
|
9
|
Saccullo E, Patamia V, Tomarchio EG, Zagni C, Floresta G, Rescifina A. Unveiling the chemistry of antibody conjugation for enhanced PET imaging: Current trends and future directions. Bioorg Chem 2025; 155:108115. [PMID: 39756200 DOI: 10.1016/j.bioorg.2024.108115] [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: 10/03/2024] [Revised: 12/23/2024] [Accepted: 12/28/2024] [Indexed: 01/07/2025]
Abstract
Positron Emission Tomography (PET) has emerged as a powerful imaging technique in molecular medicine, enabling the non-invasive visualisation and quantification of biological processes at the molecular level. Antibody-based PET imaging has recently gained prominence, offering specific targeting capabilities for various diseases. This scientific article delves into the intricate chemistry underlying antibody conjugation strategies for PET, providing a comprehensive understanding of the key principles and advancements in this rapidly evolving field. The article begins with a detailed exploration of various antibody conjugation methodologies, encompassing both covalent and non-covalent approaches. The chemical intricacies of bioconjugation reactions, such as amine and thiol chemistry, click chemistry, and bioorthogonal chemistry, are thoroughly discussed in the context of antibody modification. Additionally, the article critically analyses recent advancements in radiolabeling strategies for PET, including using radionuclides with favourable decay characteristics. This discussion covers both traditional radioisotopes and emerging alternatives, demonstrating their potential to raise the effectiveness of PET imaging agents based on antibodies. Ultimately, this article aims to contribute to the ongoing efforts to advance the field toward more effective diagnostic tools for personalized medicine.
Collapse
Affiliation(s)
- Erika Saccullo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy; Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy
| | - Vincenzo Patamia
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy.
| | - Elisabetta Grazia Tomarchio
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy; Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy
| | - Chiara Zagni
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy
| | - Giuseppe Floresta
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy.
| | - Antonio Rescifina
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
10
|
He Y, Tian R, Xu D, Wu Y, Rina S, Chen T, Guan Y, Xie T, Ying T, Xie F, Han J. Preclinical evaluation and pilot clinical study of [ 68Ga]Ga-NOTA-H006 for non-invasive PET imaging of 5T4 oncofetal antigen. Eur J Nucl Med Mol Imaging 2025; 52:611-622. [PMID: 39377811 DOI: 10.1007/s00259-024-06941-1] [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: 02/28/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
PURPOSE Trophoblast glycoprotein, the so-called 5T4, is an oncofetal antigen expressed in many different cancers. However, no 5T4-specific radioligand is employed in the clinic for non-invasive diagnosis. Thus, the aim of the current study was to develop a PET radiotracer for imaging 5T4 expression in preclinical and clinical stages. METHODS A VHH library was constructed by camel immunization. The specificity of the VHHs toward 5T4 antigen was screened through phage display biopanning and periplasmic extract enzyme-linked immunosorbent assay. 1,4,7-Triazacyclononane-1,4,7-triacetate acid (NOTA) derivative was conjugated to the selected VHH. After radiolabeling, microPET/CT and ex vivo biodistribution were conducted using BxPC-3 and MDA-MB-468 tumor-bearing mice. Cold VHH was co-injected with the tracer to challenge its binding in vivo. For the pilot clinical study, PET/CT images were acquired at 1 h after injection of tracer in patients with pathologically confirmed primary and metastatic tumors. RESULTS A library with a capacity of 1.2 × 1012 colony-forming units was constructed after successful camel immunization. Nb1-40 with a median effect concentration of 0.43 nM was selected. After humanization, the resulting H006 maintained a high affinity towards 5T4. [68Ga]Ga-NOTA-H006 with the molar activities of 6.48-54.2 GBq/µmol was prepared with high radiochemical purity (> 98%). Using [68Ga]Ga-NOTA-H006, microPET/CT revealed a clear visualization of 5T4 expression in BxPC-3 tumor-bearing mice. Ex vivo biodistribution showed that the highest tumor-to-blood ratio (∼ 3-fold) and tumor-to-muscle ratio (∼ 5-fold) were achieved at 60 min post-injection. Co-injection of the cold H006 at a dose of 1.5 mg/kg significantly reduced the tumor uptake (p < 0.0001). In the pilot clinical study, [68Ga]Ga-NOTA-H006 demonstrated its capacity to map 5T4-positive lesions in humans and yielded a mean effective dose of 3.4 × 10- 2 mSv/MBq. CONCLUSIONS [68Ga]Ga-NOTA-H006, which can visualize 5T4 expression in vivo, has been successfully developed. This opens up opportunities for non-invasively studying 5T4 expression through nuclear medicine. Further clinical investigations are warranted to explore its clinical value in disease progression and companion diagnosis.
Collapse
Affiliation(s)
- Yingfang He
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China
| | - Ruhua Tian
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Dong Xu
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanfei Wu
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200233, China
| | - Sa Rina
- Huahe Pharmaceutical Co., Ltd, Shanghai, China
| | - Tengxiang Chen
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200233, China
| | - Tianwu Xie
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China
| | - Tianlei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200233, China.
| | - Junbin Han
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China.
| |
Collapse
|
11
|
Dioury F, San C, Gnanalingam G, Henoumont C, Rousselin Y, Haouz A, Shepard W, Hosten B, Vijayakumar K, Laurent S, Port M. Bifunctional Hexadentate Pyclen-Based Chelating Agent for Mild Radiofluorination in Aqueous Solution at Room Temperature with a Ga- 18F Ternary Complex. Chemistry 2024; 30:e202403358. [PMID: 39331479 PMCID: PMC11618045 DOI: 10.1002/chem.202403358] [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: 09/20/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 09/29/2024]
Abstract
Positron Emission Tomography (PET) is used in oncology for tumor diagnosis, commonly relying on fluorine-18 (18F) emission detection. The conventional method of 18F incorporation on to probes by covalent bonding is harsh for sensitive biomolecules, which are nonetheless compounds of choice for the development of targeted probes. This study explores gallium-18F (Ga18F) coordination, a milder alternative method occurring in aqueous media at the final stage of radiosyntheses. Pyclen-based chelating agents were proposed to capture (GaF) species at room temperature and pH≥5 making the radiofluorination process compatible with heat- and acid-sensitive biomolecules. Highly promising results were obtained with the PC2A-based chelating agent LH2 derived from the new bifunctional PC2A-OAE-NCS compound. The solid-state structure of GaF(L) was elucidated by X-ray diffraction and revealed an unconventional heptacoordination of Ga(III). A high radiochemical conversion (RCC) of 86 % was achieved at room temperature, in water at pH 5 within 20 minutes. Stability studies showed the robustness of the GaF(L) complex in aqueous media for at least one day and at least one hour for the radiolabeled analog Ga18F(L). These findings demonstrated that PC2A-based compounds are chelating agents of choice for (Ga18F) species, suggesting a real technological breakthrough for PET imaging and precision medicine.
Collapse
Affiliation(s)
- Fabienne Dioury
- Conservatoire national des arts et métiersLaboratoire Génomique, bioinformatique et chimie moléculaire (GBCM)EA 75282 rue Conté75003ParisFrance
| | - Carine San
- Conservatoire national des arts et métiersLaboratoire Génomique, bioinformatique et chimie moléculaire (GBCM)EA 75282 rue Conté75003ParisFrance
- Hôpital Saint-LouisUniversité Paris CitéInstitut de Recherche Saint-LouisUnité Claude Kellershohn1 avenue Claude Vellefaux75010ParisFrance
| | - Gayathiri Gnanalingam
- Conservatoire national des arts et métiersLaboratoire Génomique, bioinformatique et chimie moléculaire (GBCM)EA 75282 rue Conté75003ParisFrance
| | - Céline Henoumont
- Université de Mons, GeneralOrganic and Biomedical Chemistry GroupNMR and Molecular Imaging Laboratory, Mendeleev building19 avenue MaistriauB-7000MonsBelgique
| | - Yoann Rousselin
- Université de BourgogneInstitut de Chimie Moléculaire de l'Université de Bourgogne (ICMUB)UMR CNRS 63029 avenue Alain Savary21078DijonFrance
| | - Ahmed Haouz
- Institut PasteurCrystallography Platform C2RTCNRS UMR 352825-28 rue du Docteur Roux75015ParisFrance
| | - William Shepard
- Synchrotron SOLEILProxima 2A, L'Orme des MerisiersDépartementale 12891190Saint-AubinFrance
| | - Benoît Hosten
- Hôpital Saint-LouisUniversité Paris CitéInstitut de Recherche Saint-LouisUnité Claude Kellershohn1 avenue Claude Vellefaux75010ParisFrance
- Université Paris CitéINSERM UMR−S 1144Optimisation Thérapeutique en Neuropsychopharmacologie4 avenue de l'Observatoire75006ParisFrance
| | - Kamsana Vijayakumar
- Conservatoire national des arts et métiersLaboratoire Génomique, bioinformatique et chimie moléculaire (GBCM)EA 75282 rue Conté75003ParisFrance
| | - Sophie Laurent
- Université de Mons, GeneralOrganic and Biomedical Chemistry GroupNMR and Molecular Imaging Laboratory, Mendeleev building19 avenue MaistriauB-7000MonsBelgique
| | - Marc Port
- Conservatoire national des arts et métiersLaboratoire Génomique, bioinformatique et chimie moléculaire (GBCM)EA 75282 rue Conté75003ParisFrance
| |
Collapse
|
12
|
Wu Q, Shao H, Zhai W, Huang G, Liu J, Calais J, Wei W. Molecular imaging of renal cell carcinomas: ready for prime time. Nat Rev Urol 2024:10.1038/s41585-024-00962-z. [PMID: 39543358 DOI: 10.1038/s41585-024-00962-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2024] [Indexed: 11/17/2024]
Abstract
The clinical diagnosis of renal cell carcinoma (RCC) is constantly evolving. Diagnostic imaging of RCC relying on enhanced computed tomography (CT) and magnetic resonance imaging (MRI) is commonly used for renal mass characterization and assessment of tumour thrombosis, whereas pathology is the gold standard for establishing diagnosis. However, molecular imaging is rapidly improving the clinical management of RCC, particularly clear-cell RCC. Molecular imaging aids in the non-invasive visualization and characterization of specific biomarkers such as carbonic anhydrase IX and CD70 within the tumours, which help to assess tumour heterogeneity and status. Target-specific molecular imaging of RCCs will substantially improve the diagnostic landscape of RCC and will further facilitate clinical decision-making regarding initial staging and re-staging, monitoring of recurrence and metastasis, patient stratification and selection, and the prediction and evaluation of treatment responses.
Collapse
Affiliation(s)
- Qianyun Wu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hongda Shao
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Zhai
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA.
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
13
|
Huang W, Cao M, Wu Y, Zhang Y, An S, Pan X, Zhou X, Shao H, Guan Y, Huang G, Gelardi F, Chiti A, Xie F, Liu J, Wei W. Immuno-PET/CT Imaging of Trop2 with [ 18F]AlF-RESCA-T4 Differentiates Lung Cancer from Inflammation. J Nucl Med 2024:jnumed.124.268751. [PMID: 39542697 DOI: 10.2967/jnumed.124.268751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024] Open
Abstract
Immuno-PET/CT imaging, a branch of molecular imaging, can noninvasively and specifically visualize biomarker expression across the body. Trophoblast cell surface antigen 2 (Trop2) is a pan-cancer biomarker and plays a crucial role in tumorigenesis through multiple signaling pathways. The study aims to develop and translate novel Trop2 single-domain antibody (sdAb) tracers for clinical use. Methods: Two sdAbs (i.e., His-tagged T4 and His-tag-free RT4) are recombinantly expressed in Chinese hamster ovary cells. The purities and binding kinetics are determined by sodium dodecyl sulfate polyacrylamide gel electrophoresis, high-performance liquid chromatography, and surface plasmon resonance assays. The AlF restrained complexing agent (RESCA) method is applied to develop 18F-labeled sdAb tracers ([18F]AlF-RESCA-T4 and [18F]AlF-RESCA-RT4), followed by thorough preclinical imaging and blocking studies on tumor-bearing mice and a pilot clinical trial evaluating the clinical imaging safety and feasibility of [18F]AlF-RESCA-T4 immuno-PET/CT. Results: [18F]AlF-RESCA-T4 and [18F]AlF-RESCA-RT4 possess high radiochemical purities. Preclinical imaging in the T3M-4 tumor model revealed prominent uptake (percentage injected dose/g) of [18F]AlF-RESCA-T4 (11.13 ± 1.53, n = 4) and [18F]AlF-RESCA-RT4 (8.83 ± 1.22, n = 4), which were significantly reduced by coinjection of unlabeled T4 and RT4 in blocking studies. The His-tag removal strategy further optimized the probe's in vivo pharmacokinetics and reduced renal radioactivity accumulation without significantly decreasing tumor uptake. In a pilot clinical trial, [18F]AlF-RESCA-T4 immuno-PET/CT showed promising potency in annotating Trop2 expression and differentiating tumors from inflammatory diseases such as tuberculosis. Conclusion: [18F]AlF-RESCA-T4 and [18F]AlF-RESCA-RT4 can specifically annotate Trop2 expression. Clinical [18F]AlF-RESCA-T4 immuno-PET/CT imaging can screen patients for Trop2-targeted therapies and differentiate lung inflammation from cancer.
Collapse
Affiliation(s)
- Wei Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Min Cao
- Department of Thoracic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanfei Wu
- Department of Nuclear Medicine and PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - You Zhang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shuxian An
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xinbing Pan
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xinyuan Zhou
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hongda Shao
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yihui Guan
- Department of Nuclear Medicine and PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Fabrizia Gelardi
- Università Vita-Salute San Raffaele, Milan, Italy; and
- Nuclear Medicine Department, IRCCS San Raffaele, Milano, Italy
| | - Arturo Chiti
- Università Vita-Salute San Raffaele, Milan, Italy; and
- Nuclear Medicine Department, IRCCS San Raffaele, Milano, Italy
| | - Fang Xie
- Department of Nuclear Medicine and PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China;
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China;
| |
Collapse
|
14
|
Lechi F, Eriksson J, Odell LR, Wegrzyniak O, Löfblom J, Frejd FY, Zhang B, Eriksson O. Optimized method for fluorine-18 radiolabeling of Affibody molecules using RESCA. EJNMMI Radiopharm Chem 2024; 9:73. [PMID: 39460878 PMCID: PMC11512968 DOI: 10.1186/s41181-024-00304-9] [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: 08/19/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND In recent years, the interest in Al[18F]F as a labeling agent for Positron Emission Tomography (PET) radiotracers has risen, as it allows for fast and efficient fluorine-18 labeling by harnessing chelation chemistry. The introduction of Restrained Complexing Agent (RESCA) as a chelator has also shown that chelator-based radiolabeling reactions can be performed in mild conditions, making the radiolabeling process attractively more facile than most conventional radiofluorination methods. The aim of the study was to establish optimized conditions for Al[18F]F labeling of Affibody molecules using RESCA as a complexing agent, using Z09591 and Z0185, two Affibody proteins targeting PDGFRβ and TNFα, respectively, as model compounds. RESULTS The Al[18F]F labeling of RESCA-conjugated Z09591 was tested at different temperatures (rt to 60 °C) and with varying reaction times (12 to 60 min), and optimal conditions were then implemented on RESCA-Z0185. The optimized synthesis method was: 1.5-2.5 GBq of cyclotron produced fluorine-18 were trapped on a QMA cartridge and eluted with saline solution to react with 12 nmol of AlCl3 and form Al[18F]F. The respective RESCA-conjugated Affibody molecule (14 nmol) in NaOAc solution was added to the Al[18F]F solution and left to react at 60 °C for 12 min. The mixture was purified on a NAP5 size exclusion column and then analyzed by HPLC. The entire process took approximately 35 min, was highly reproducible, indicating the efficiency and reliability of the method. The labeled compounds demonstrated retained biological function for their respective targets after purification. CONCLUSIONS We present a general and optimized method for Al[18F]F labeling of RESCA-conjugated Affibody molecules, which can be widely applied to this class of peptide-based imaging agents. Moreover, radiochemical yields were improved when the labeling was conducted at 37 °C or above. In vitro and in vivo assessment of the respective tracers was promising, showing retained binding capacity as well as moderate defluorination, which is usually regarded as a potential downside for RESCA-conjugated tracers.
Collapse
Affiliation(s)
- Francesco Lechi
- Department of Medicinal Chemistry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Jonas Eriksson
- Department of Medicinal Chemistry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- PET center, Uppsala University Hospital, Uppsala, Sweden
| | - Luke R Odell
- Department of Medicinal Chemistry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Olivia Wegrzyniak
- Department of Medicinal Chemistry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - John Löfblom
- Division of Protein Engineering, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Fredrik Y Frejd
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Affibody AB, Solna, Sweden
| | - Bo Zhang
- Department of Medicinal Chemistry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Olof Eriksson
- Department of Medicinal Chemistry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
15
|
Sihver W, Walther M, Ullrich M, Nitt-Weber AK, Böhme J, Reissig F, Saager M, Zarschler K, Neuber C, Steinbach J, Kopka K, Pietzsch HJ, Wodtke R, Pietzsch J. Cyclohexanediamine Triazole (CHDT) Functionalization Enables Labeling of Target Molecules with Al 18F/ 68Ga/ 111In. Bioconjug Chem 2024; 35:1402-1416. [PMID: 39185789 PMCID: PMC11417994 DOI: 10.1021/acs.bioconjchem.4c00313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 08/27/2024]
Abstract
The Al18F-labeling approach offers a one-step access to radiofluorinated biomolecules by mimicking the labeling process for radiometals. Although these labeling conditions are considered to be mild compared to classic radiofluorinations, improvements of the chelating units have led to the discovery of (±)-H3RESCA, which allows Al18F-labeling already at ambient temperature. While the suitability of (±)-H3RESCA for functionalization and radiofluorination of proteins is well established, its use for small molecules or peptides is less explored. Herein, we advanced this acyclic pentadentate ligand by introducing an alkyne moiety for the late-stage functionalization of biomolecules via click chemistry. We show that in addition to Al18F-labeling, the cyclohexanediamine triazole (CHDT) moiety allows stable complexation of 68Ga and 111In. Three novel CHDT-functionalized PSMA inhibitors were synthesized and their Al18F-, 68Ga-, and 111In-labeled analogs were subjected to a detailed in vitro radiopharmacological characterization. Stability studies in vitro in human serum revealed among others a high kinetic inertness of all radiometal complexes. Furthermore, the Al18F-labeled PSMA ligands were characterized for their biodistribution in a LNCaP derived tumor xenograft mouse model by PET imaging. One radioligand, Al[18F]F-CHDT-PSMA-1, bearing a small azidoacetyl linker at the glutamate-urea-lysine motif, provided an in vivo performance comparable to that of [18F]PSMA-1007 but with even higher tumor-to-blood and tumor-to-muscle ratios at 120 min p.i. Overall, our results highlight the suitability of the novel CHDT moiety for functionalization and radiolabeling of small molecules or peptides with Al18F, 68Ga, and 111In and the triazole ring seems to entail favorable pharmacokinetic properties for molecular imaging purposes.
Collapse
Affiliation(s)
- Wiebke Sihver
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, Bautzner
Landstraße 400, 01328 Dresden, Germany
| | - Martin Walther
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, Bautzner
Landstraße 400, 01328 Dresden, Germany
| | - Martin Ullrich
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, Bautzner
Landstraße 400, 01328 Dresden, Germany
| | - Anne-Kathrin Nitt-Weber
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, Bautzner
Landstraße 400, 01328 Dresden, Germany
| | - Jenny Böhme
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, Bautzner
Landstraße 400, 01328 Dresden, Germany
| | - Falco Reissig
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, Bautzner
Landstraße 400, 01328 Dresden, Germany
| | - Magdalena Saager
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, Bautzner
Landstraße 400, 01328 Dresden, Germany
| | - Kristof Zarschler
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, Bautzner
Landstraße 400, 01328 Dresden, Germany
| | - Christin Neuber
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, Bautzner
Landstraße 400, 01328 Dresden, Germany
| | - Jörg Steinbach
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, Bautzner
Landstraße 400, 01328 Dresden, Germany
| | - Klaus Kopka
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, Bautzner
Landstraße 400, 01328 Dresden, Germany
- Technische
Universität Dresden, School of Science,
Faculty of Chemistry and Food Chemistry, Mommsenstraße 4, 01069 Dresden, Germany
| | - Hans-Jürgen Pietzsch
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, Bautzner
Landstraße 400, 01328 Dresden, Germany
- Technische
Universität Dresden, School of Science,
Faculty of Chemistry and Food Chemistry, Mommsenstraße 4, 01069 Dresden, Germany
| | - Robert Wodtke
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, Bautzner
Landstraße 400, 01328 Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, Bautzner
Landstraße 400, 01328 Dresden, Germany
- Technische
Universität Dresden, School of Science,
Faculty of Chemistry and Food Chemistry, Mommsenstraße 4, 01069 Dresden, Germany
| |
Collapse
|
16
|
Basuli F, Shi J, Lindberg E, Fayn S, Lee W, Ho M, Hammoud DA, Cheloha RW, Swenson RE, Escorcia FE. Sortase-Mediated Site-Specific Conjugation to Prepare Fluorine-18-Labeled Nanobodies. Bioconjug Chem 2024; 35:1335-1342. [PMID: 39172920 DOI: 10.1021/acs.bioconjchem.4c00264] [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] [Indexed: 08/24/2024]
Abstract
Single-domain antibodies, or nanobodies (Nbs), are promising biomolecules for use in molecular imaging due to their excellent affinity, specificity, and fast clearance from the blood. Given their short blood half-life, pairing Nbs with short-lived imaging radioisotopes is desirable. Because fluorine-18 (18F) is routinely used for clinical imaging, it is an attractive radioisotope for Nbs. We report a novel sortase-based, site-specific 18F-labeling method applied to three nanobodies. Labeled nanobodies were synthesized either by a two-step indirect radiolabeling method in one pot or by a one-step direct labeling method using a sortase-mediated conjugation of either the radiolabeled chelator (H-GGGK((±)-Al[18F]FH3RESCA)-NH2) or the unlabeled chelator (H-GGGK((±)-H3RESCA)-NH2) followed by labeling with Al[18F]F, respectively. The overall radiochemical yields were 15-43% (n = 22, decay-corrected) in 70 min (indirect labeling) and 23-58% (n = 12, decay-corrected) in 50 min (direct labeling). The radiochemical purities of the labeled nanobodies prepared by both methods were >98% with a specific activity of 400-600 Ci/mmol (n = 22) for each of the three Nbs tested and exhibited excellent stability profiles under physiological conditions. This simple, site-specific, reproducible, and generalizable 18F-labeling method to prepare nanobodies (Nb-Al[18F]F-RESCA) or other low molecular weight biomolecules can easily be adopted in various settings for preclinical and clinical studies.
Collapse
Affiliation(s)
- Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, Maryland 20892-0001, United States
| | - Jianfeng Shi
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, Maryland 20892-0001, United States
| | - Eric Lindberg
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, Maryland 20892-0001, United States
| | - Stanley Fayn
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford OX3 7DQ, U.K
| | - Woonghee Lee
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Mitchell Ho
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Dima A Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health (NIH), Bethesda, Maryland 20892, United States
| | - Ross W Cheloha
- Chemical Biology in Signaling Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Rolf E Swenson
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, Maryland 20892-0001, United States
| | - Freddy E Escorcia
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
17
|
Zeven K, Lauwers Y, De Mey L, Debacker JM, De Pauw T, De Groof TWM, Devoogdt N. Advancements in nuclear imaging using radiolabeled nanobody tracers to support cancer immunotherapy. IMMUNOTHERAPY ADVANCES 2024; 4:ltae006. [PMID: 39281708 PMCID: PMC11402390 DOI: 10.1093/immadv/ltae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/23/2024] [Indexed: 09/18/2024] Open
Abstract
The evolving landscape of cancer immunotherapy has revolutionized cancer treatment. However, the dynamic tumor microenvironment has led to variable clinical outcomes, indicating a need for predictive biomarkers. Noninvasive nuclear imaging, using radiolabeled modalities, has aided in patient selection and monitoring of their treatment response. This approach holds promise for improving diagnostic accuracy, providing a more personalized treatment regimen, and enhancing the clinical response. Nanobodies or single-domain antibodies, derived from camelid heavy-chain antibodies, allow early timepoint detection of targets with high target-to-background ratios. To date, a plethora of nanobodies have been developed for nuclear imaging of tumor-specific antigens, immune checkpoints, and immune cells, both at a preclinical and clinical level. This review comprehensively outlines the recent advancements in nanobody-based nuclear imaging, both on preclinical and clinical levels. Additionally, the impact and expected future advancements on the use of nanobody-based radiopharmaceuticals in supporting cancer diagnosis and treatment follow-up are discussed.
Collapse
Affiliation(s)
- Katty Zeven
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Yoline Lauwers
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Lynn De Mey
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Jens M Debacker
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Tessa De Pauw
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Timo W M De Groof
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Nick Devoogdt
- Molecular Imaging and Therapy Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
18
|
Dierick H, Navarro L, Ceuppens H, Ertveldt T, Pombo Antunes AR, Keyaerts M, Devoogdt N, Breckpot K, D'Huyvetter M, Lahoutte T, Caveliers V, Bridoux J. Generic semi-automated radiofluorination strategy for single domain antibodies: [ 18F]FB-labelled single domain antibodies for PET imaging of fibroblast activation protein-α or folate receptor-α overexpression in cancer. EJNMMI Radiopharm Chem 2024; 9:54. [PMID: 39048805 PMCID: PMC11269545 DOI: 10.1186/s41181-024-00286-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Radiofluorination of single domain antibodies (sdAbs) via N-succinimidyl-4-[18F]fluorobenzoate ([18F]SFB) has shown to be a promising strategy in the development of sdAb-based PET tracers. While automation of the prosthetic group (PG) [18F]SFB production, has been successfully reported, no practical method for large scale sdAb labelling has been reported. Therefore, we optimized and automated the PG production, enabling a subsequently efficient manual conjugation reaction to an anti-fibroblast activation protein (FAP)-α sdAb (4AH29) and an anti-folate receptor (FR)-α sdAb (2BD42). Both the alpha isoform of FAP and the FR are established tumour markers. FAP-α is known to be overexpressed mainly by cancer-associated fibroblasts in breast, ovarian, and other cancers, while its expression in normal tissues is low or undetectable. FR-α has an elevated expression in epithelial cancers, such as ovarian, brain and lung cancers. Non-invasive imaging techniques, such as PET-imaging, using tracers targeting specific tumour markers can provide molecular information over both the tumour and its environment, which aides in the diagnosis, therapy selection and assessment of the cancer treatment. RESULTS [18F]SFB was synthesized using a fully automated three-step, one-pot reaction. The total procedure time was 54 min and results in [18F]SFB with a RCP > 90% and a RCY d.c. of 44 ± 4% (n = 13). The manual conjugation reaction after purification produced [18F]FB-sdAbs with a RCP > 95%, an end of synthesis activity > 600 MBq and an apparent molar activity > 10 GBq/µmol. Overall RCY d.c., corrected to the trapping of [18F]F- on the QMA, were 9% (n = 1) and 5 ± 2% (n = 3) for [18F]FB-2BD42 and [18F]FB-4AH29, respectively. CONCLUSION [18F]SFB synthesis was successfully automated and upscaled on a Trasis AllInOne module. The anti-hFAP-α and anti-hFR-α sdAbs were radiofluorinated, yielding similar RCYs d.c. and RCPs, showing the potential of this method as a generic radiofluorination strategy for sdAbs. The radiofluorinated sdAbs showed a favourable biodistribution pattern and are attractive for further characterization as new PET tracers for FAP-α and FR-α imaging.
Collapse
Affiliation(s)
- Herlinde Dierick
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103. Building K., 1090, Brussels, Belgium.
- Nuclear Medicine Department, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090, Brussels, Belgium.
| | - Laurent Navarro
- Precirix NV, Burgemeester Etienne Demunterlaan 3, 1090, Brussels, Belgium
| | - Hannelore Ceuppens
- Laboratory for Molecular and Cellular Therapy (LCMT), Department of Biomedical Sciences, Translational Oncology Research Center, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103. Building E, 1090, Brussels, Belgium
| | - Thomas Ertveldt
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103. Building K., 1090, Brussels, Belgium
- Laboratory for Molecular and Cellular Therapy (LCMT), Department of Biomedical Sciences, Translational Oncology Research Center, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103. Building E, 1090, Brussels, Belgium
| | | | - Marleen Keyaerts
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103. Building K., 1090, Brussels, Belgium
| | - Nick Devoogdt
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103. Building K., 1090, Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy (LCMT), Department of Biomedical Sciences, Translational Oncology Research Center, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103. Building E, 1090, Brussels, Belgium
| | - Matthias D'Huyvetter
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103. Building K., 1090, Brussels, Belgium
- Precirix NV, Burgemeester Etienne Demunterlaan 3, 1090, Brussels, Belgium
| | - Tony Lahoutte
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103. Building K., 1090, Brussels, Belgium
- Nuclear Medicine Department, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Vicky Caveliers
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103. Building K., 1090, Brussels, Belgium
- Nuclear Medicine Department, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Jessica Bridoux
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103. Building K., 1090, Brussels, Belgium
| |
Collapse
|
19
|
Wu Q, Wu Y, Zhang Y, Guan Y, Huang G, Xie F, Liu J, Zhai W, Wei W. ImmunoPET/CT imaging of clear cell renal cell carcinoma with [ 18F]RCCB6: a first-in-human study. Eur J Nucl Med Mol Imaging 2024; 51:2444-2457. [PMID: 38480552 DOI: 10.1007/s00259-024-06672-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 03/05/2024] [Indexed: 06/15/2024]
Abstract
PURPOSE The cluster of differentiation (CD70) is a potential biomarker of clear cell renal cell carcinoma (ccRCC). This study aims to develop CD70-targeted immuno-positron emission tomography/computed tomography (immunoPET/CT) imaging tracers and explore the diagnostic value in preclinical studies and the potential value in detecting metastases in ccRCC patients. METHODS Four novel CD70-specific single-domain antibodies (sdAbs) were produced and labelled with 18F by the aluminium fluoride restrained complexing agent (AlF-RESCA) method to develop radiotracers. The visualisation properties of the tracers were evaluated in a subcutaneous ccRCC patient-derived xenograft (PDX) model. In a registered prospective clinical trial (NCT06148220), six patients with pathologically confirmed RCC were included and underwent immunoPET/CT examination exploiting one of the developed tracers (i.e., [18F]RCCB6). RESULTS We engineered four sdAbs (His-tagged RCCB3 and RCCB6, His-tag-free RB3 and RB6) specifically targeting recombinant human CD70 without cross-reactivity to murine CD70. ImmunoPET/CT imaging with [18F]RCCB3 and [18F]RCCB6 demonstrated a high tumour-to-background ratio in a subcutaneous ccRCC PDX model, with the latter showing better diagnostic potential supported by higher tumour uptake and lower bone accumulation. In comparison, [18F]RB6, developed by sequence optimisation, has significantly lower kidney accumulation than that of [18F]RCCB6. In a pilot translational study, [18F]RCCB6 immunoPET/CT displayed ccRCC metastases in multiple patients and demonstrated improved imaging contrast and diagnostic value than 18F-FDG PET/CT in a patient with ccRCC. CONCLUSION The work successfully developed a series of CD70-targeted immunoPET/CT imaging tracers. Of them, [18F]RCCB6 clearly and specifically identified inoculated ccRCCs in preclinical studies. Clinical translation of [18F]RCCB6 suggests potential for identifying recurrence and/or metastasis in ccRCC patients.
Collapse
Affiliation(s)
- Qianyun Wu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Yanfei Wu
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - You Zhang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China.
| | - Wei Zhai
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China.
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China.
| |
Collapse
|
20
|
Iannone MN, Valtorta S, Stucchi S, Altomonte S, Turolla EA, Vino E, Rainone P, Zecca V, Lo Dico A, Maspero M, Figini M, Bellone M, Ciceri S, Colombo D, Chinello C, Pagani L, Moresco RM, Todde S, Ferraboschi P. Automated radiosynthesis and preclinical evaluation of two new PSMA-617 derivatives radiolabelled via [ 18F]AlF 2+ method. EJNMMI Radiopharm Chem 2024; 9:50. [PMID: 38904859 PMCID: PMC11192711 DOI: 10.1186/s41181-024-00280-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 06/10/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND In the last decade the development of new PSMA-ligand based radiopharmaceuticals for the imaging and therapy of prostate cancer has been a highly active and important area of research. The most promising derivative in terms of interaction with the antigen and clinical properties has been found to be "PSMA-617", and its lutetium-177 radiolabelled version has recently been approved by EU and USA regulatory agencies for therapeutic purposes. For the above reasons, the development of new derivatives of PSMA-617 radiolabelled with fluorine-18 may still be of great interest. This paper proposes the comparison of two different PSMA-617 derivatives functionalized with NODA and RESCA chelators, respectively, radiolabelled via [18F]AlF2+ complexation. RESULTS The organic synthesis of two PSMA-617 derivatives and their radiolabelling via [18F]AlF2+ complexation resulted to proceed efficiently and successfully. Moreover, stability in solution and in plasma has been evaluated. The whole radiosynthesis procedure has been fully automated, and the final products have been obtained with radiochemical yield and purity potentially suitable for clinical studies. The biodistribution of the two derivatives was performed both in prostate cancer and glioma tumour models. Compared with the reference [18F]F-PSMA-1007 and [18F]F-PSMA-617-RESCA, [18F]F-PSMA-617-NODA derivative showed a higher uptake in both tumors, faster clearance in non-target organs, and lower uptake in salivary glands. CONCLUSION PSMA-617 NODA and RESCA derivatives were radiolabelled successfully via [18F]AlF2+ chelation, the former being more stable in solution and human plasma. Moreover, preclinical biodistribution studies showed that [18F]F-PSMA-617-NODA might be of potential interest for clinical applications.
Collapse
Affiliation(s)
| | - Silvia Valtorta
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Segrate, Italy
- NBFC, National Biodiversity Future Center, Palermo, Italy
- Department of Nuclear Medicine, San Raffaele Scientific Institute, IRCCS, Milan, Italy
| | - Stefano Stucchi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Stefano Altomonte
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Elia Anna Turolla
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Elisa Vino
- Tecnomed Foundation, University of Milano-Bicocca, Monza, Italy
| | - Paolo Rainone
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Segrate, Italy
- Department of Nuclear Medicine, San Raffaele Scientific Institute, IRCCS, Milan, Italy
| | - Valentina Zecca
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Segrate, Italy
- Department of Nuclear Medicine, San Raffaele Scientific Institute, IRCCS, Milan, Italy
| | - Alessia Lo Dico
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Segrate, Italy
- NBFC, National Biodiversity Future Center, Palermo, Italy
| | - Marco Maspero
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Segrate, Italy
- Department of Nuclear Medicine, San Raffaele Scientific Institute, IRCCS, Milan, Italy
| | - Mariangela Figini
- ANP2, Department of Advanced Diagnostics, Fondazione IRCCS, Istituto Nazionale Dei Tumori, Milan, Italy
| | - Matteo Bellone
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, IRCCS, Milan, Italy
| | - Samuele Ciceri
- Department of Medical Biotechnology and Translational Medicine, University of Milano, Milan, Italy
| | - Diego Colombo
- Department of Medical Biotechnology and Translational Medicine, University of Milano, Milan, Italy
| | - Clizia Chinello
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Lisa Pagani
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Rosa Maria Moresco
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Segrate, Italy
- Department of Nuclear Medicine, San Raffaele Scientific Institute, IRCCS, Milan, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Sergio Todde
- Tecnomed Foundation, University of Milano-Bicocca, Monza, Italy
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Segrate, Italy
| | - Patrizia Ferraboschi
- Department of Medical Biotechnology and Translational Medicine, University of Milano, Milan, Italy
| |
Collapse
|
21
|
Wegrzyniak O, Lechi F, Mitran B, Cheung P, Bitzios A, Persson J, Löfblom J, Nordström H, Eriksson J, Frejd FY, Korsgren O, Zhang B, Eriksson O. Non-invasive PET imaging of liver fibrogenesis using a RESCA-conjugated Affibody molecule. iScience 2024; 27:109688. [PMID: 38660405 PMCID: PMC11039342 DOI: 10.1016/j.isci.2024.109688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 02/02/2024] [Accepted: 04/05/2024] [Indexed: 04/26/2024] Open
Abstract
Non-invasive assessment of fibrogenic activity, rather than fibrotic scars, could significantly improve the management of fibrotic diseases and the development of anti-fibrotic drugs. This study explores the potential of an Affibody molecule (Z09591) labeled with the Al(18)F-restrained complexing agent (RESCA) method as a tracer for the non-invasive detection of fibrogenic cells. Z09591 was functionalized with the RESCA chelator for direct labeling with [18F]AlF. In vivo positron emission tomography/magnetic resonance imaging scans on U-87 tumor-bearing mice exhibited high selectivity of the resulting radiotracer, [18F]AlF-RESCA-Z09591, for platelet-derived growth factor receptor β (PDGFRβ), with minimal non-specific background uptake. Evaluation in a mouse model with carbon tetrachloride-induced fibrotic liver followed by a disease regression phase, revealed the radiotracer's high affinity and specificity for fibrogenic cells in fibrotic livers (standardized uptake value [SUV] 0.43 ± 0.05), with uptake decreasing during recovery (SUV 0.29 ± 0.03) (p < 0.0001). [18F]AlF-RESCA-Z09591 accurately detects PDGFRβ, offering non-invasive assessment of fibrogenic cells and promising applications in precise liver fibrogenesis diagnosis, potentially contributing significantly to anti-fibrotic drug development.
Collapse
Affiliation(s)
- Olivia Wegrzyniak
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Francesco Lechi
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Bogdan Mitran
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
- Antaros Medical AB, Mölndal, Sweden
| | - Pierre Cheung
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Athanasios Bitzios
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Jonas Persson
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
- Department of Protein Science, Division of Protein Engineering, KTH Royal Institute of Technology, Stockholm, Sweden
| | - John Löfblom
- Department of Protein Science, Division of Protein Engineering, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Helena Nordström
- Science for Life Laboratory, Drug Discovery & Development Platform, Department of Chemistry-BMC, Uppsala University, Uppsala, Sweden
| | - Jonas Eriksson
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
- PET Center, Uppsala University Hospital, Uppsala, Sweden
| | - Fredrik Y. Frejd
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Affibody AB, Solna, Sweden
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Bo Zhang
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Olof Eriksson
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
- Antaros Medical AB, Mölndal, Sweden
| |
Collapse
|
22
|
Gao X, Wang Q, Yang X, Fang J, Li H, Xi H, Lin J, Qiu L. Legumain-Triggered Macrocyclization of Radiofluorinated Tracer for Enhanced PET Imaging. Bioconjug Chem 2024; 35:665-673. [PMID: 38598424 DOI: 10.1021/acs.bioconjchem.4c00128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Enhancing the accumulation and retention of small-molecule probes in tumors is an important way to achieve accurate cancer diagnosis and therapy. Enzyme-stimulated macrocyclization of small molecules possesses great potential for enhanced positron emission tomography (PET) imaging of tumors. Herein, we reported an 18F-labeled radiotracer [18F]AlF-RSM for legumain detection in vivo. The tracer was prepared by a one-step aluminum-fluoride-restrained complexing agent ([18F]AlF-RESCA) method with high radiochemical yield (RCY) (88.35 ± 3.93%) and radiochemical purity (RCP) (>95%). More notably, the tracer can be transformed into a hydrophobic macrocyclic molecule under the joint action of legumain and reductant. Simultaneously, the tracer could target legumain-positive tumors and enhance accumulation and retention in tumors, resulting in the amplification of PET imaging signals. The enhancement of radioactivity enables PET imaging of legumain activity with high specificity. We envision that, by combining this highly efficient 18F-labeled strategy with our intramolecular macrocyclization reaction, a range of radiofluorinated tracers can be designed for tumor PET imaging and early cancer diagnosis in the future.
Collapse
Affiliation(s)
- Xiaoqing Gao
- School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China
| | - Qianhui Wang
- School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China
| | - Xiaofeng Yang
- School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China
| | - Jing Fang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China
| | - Huirong Li
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China
| | - Hongjie Xi
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China
| | - Jianguo Lin
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China
| | - Ling Qiu
- School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China
| |
Collapse
|
23
|
Sadasivam P, Khanapur S, Hartimath SV, Ramasamy B, Cheng P, Feng CZ, Green D, Davis C, Goggi JL, Robins EG, Yan R. Arginine-Selective Bioconjugation Reagent for Effective 18F-labeling of Native Proteins. J Med Chem 2024; 67:5064-5074. [PMID: 38480493 PMCID: PMC10982996 DOI: 10.1021/acs.jmedchem.4c00154] [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: 01/18/2024] [Revised: 02/28/2024] [Accepted: 03/01/2024] [Indexed: 04/04/2024]
Abstract
Protein-based 18F-PET tracers offer new possibilities in early disease detection and personalized medicine. Their development relies heavily on the availability and effectiveness of 18F-prosthetic groups. We prepared and evaluated a novel arginine-selective prosthetic group, 4-[18F]fluorophenylglyoxal ([18F]FPG). [18F]FPG was radiosynthesized by a one-pot, two-step procedure with a non-decay-corrected (n.d.c.) isolated radiochemical yield (RCY) of 41 ± 8% (n = 10). [18F]FPG constitutes a generic tool for 18F-labeling of various proteins, including human serum albumin (HSA), ubiquitin, interleukin-2, and interleukin-4 in ∼30-60% n.d.c. isolated RCYs. [18F]FPG conjugation with arginine residues is highly selective, even in the presence of a large excess of lysine, cysteine, and histidine. [18F]FPG protein conjugates are able to preserve the binding affinity of the native proteins while also demonstrating excellent in vivo stability. The [18F]FPG-HSA conjugate has prolonged blood retention, which can be applied as a potential blood pool PET imaging agent. Thus, [18F]FPG is an arginine-selective bioconjugation reagent that can be effectively used for the development of 18F-labeled protein radiopharmaceuticals.
Collapse
Affiliation(s)
- Pragalath Sadasivam
- School
of Biomedical Engineering and Imaging Sciences, Department of Imaging
Chemistry and Biology, King’s College, London SE1 7EH, U.K.
- Institute
of Bioengineering and Bioimaging, Agency
for Science, Technology, and Research (A* STAR), 11 Biopolis Way, #01-02 Helios, Singapore 138667, Singapore
- Clinical
Imaging Research Centre, 14 Medical Drive, #B01-01 Centre for Translational
Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Minerva
Imaging ApS, Lyshøjvej
21, Ølstykke 3650, Denmark
| | - Shivashankar Khanapur
- Institute
of Bioengineering and Bioimaging, Agency
for Science, Technology, and Research (A* STAR), 11 Biopolis Way, #01-02 Helios, Singapore 138667, Singapore
| | - Siddesh V. Hartimath
- Institute
of Bioengineering and Bioimaging, Agency
for Science, Technology, and Research (A* STAR), 11 Biopolis Way, #01-02 Helios, Singapore 138667, Singapore
| | - Boominathan Ramasamy
- Institute
of Bioengineering and Bioimaging, Agency
for Science, Technology, and Research (A* STAR), 11 Biopolis Way, #01-02 Helios, Singapore 138667, Singapore
| | - Peter Cheng
- Institute
of Bioengineering and Bioimaging, Agency
for Science, Technology, and Research (A* STAR), 11 Biopolis Way, #01-02 Helios, Singapore 138667, Singapore
| | - Chin Zan Feng
- Institute
of Bioengineering and Bioimaging, Agency
for Science, Technology, and Research (A* STAR), 11 Biopolis Way, #01-02 Helios, Singapore 138667, Singapore
| | - David Green
- Clinical
Imaging Research Centre, 14 Medical Drive, #B01-01 Centre for Translational
Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Christopher Davis
- School
of Biomedical Engineering and Imaging Sciences, Department of Imaging
Chemistry and Biology, King’s College, London SE1 7EH, U.K.
| | - Julian L. Goggi
- Institute
of Bioengineering and Bioimaging, Agency
for Science, Technology, and Research (A* STAR), 11 Biopolis Way, #01-02 Helios, Singapore 138667, Singapore
- Minerva
Imaging ApS, Lyshøjvej
21, Ølstykke 3650, Denmark
| | - Edward G. Robins
- Institute
of Bioengineering and Bioimaging, Agency
for Science, Technology, and Research (A* STAR), 11 Biopolis Way, #01-02 Helios, Singapore 138667, Singapore
- Clinical
Imaging Research Centre, 14 Medical Drive, #B01-01 Centre for Translational
Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Molecular
Imaging and Therapy Research Unit, South Australian Health, and Medical
Research Institute (SAHMRI), North Terrace, Adelaide, SA 5000, Australia
- Adelaide
Medical School, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace & George Street, Adelaide, SA 5000, Australia
| | - Ran Yan
- School
of Biomedical Engineering and Imaging Sciences, Department of Imaging
Chemistry and Biology, King’s College, London SE1 7EH, U.K.
| |
Collapse
|
24
|
Liu J, Guo X, Wen L, Wang L, Liu F, Song G, Zhu H, Zhou N, Yang Z. Comparison of renal clearance of [ 18F]AlF-RESCA-HER2-BCH and [ 18F]AlF-NOTA-HER2-BCH in mice and breast cancer patients. Eur J Nucl Med Mol Imaging 2023; 50:2775-2786. [PMID: 37093312 DOI: 10.1007/s00259-023-06232-1] [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: 02/16/2023] [Accepted: 04/11/2023] [Indexed: 04/25/2023]
Abstract
PURPOSE A novel HER2 affibody-based molecular probe, [18F]AlF-RESCA-HER2-BCH, was developed for reducing renal uptake, evaluated, and compared with [18F]AlF-NOTA-HER2-BCH. METHODS In preclinical studies, micro-PET/CT was performed using HER2-positive gastric cancer patient-derived xenografts (PDX) model at 0.5-1 (dynamic), 2, 4, and 6 h post-injection. For blocking experiment, 0.5 mg cold affibody was co-injected with probes. Biodistribution were performed on HER2-positive PDX models at 2 h post-injection. For clinical study, PET/CT images were acquired at 2 h and 4 h after injection of 231.29 ± 17.77 MBq [18F]AlF-NOTA-HER2-BCH or [18F]AlF-RESCA-HER2-BCH in five breast cancer patients (4 HER2-positive and 1 HER2-low). Standardized uptake values (SUVs) were measured in tumors and source-organs for semi-quantitative analysis. The OLINDA/EXM software (version 1.2) was used to calculate the radiation doses. RESULTS [18F]AlF-NOTA-HER2-BCH and [18F]AlF-RESCA-HER2-BCH were stably labeled with [18F]F, with high binding specificity and affinity to HER2. Micro-PET/CT of both tracers could clearly visualize HER2-positive PDX tumors with high uptake of 16.24 ± 1.74% ID/g and 14.39 ± 2.45% ID/g at 2 h post-injection. The renal accumulation of [18F]AlF-RESCA-HER2-BCH was significantly lower than that of [18F]AlF-NOTA-HER2-BCH (5.16 ± 0.22% ID/g vs. 158.73 ± 5.44% ID/g at 2 h, p < 0.0001). In the clinical study, both [18F]AlF-NOTA-HER2-BCH and [18F]AlF-RESCA-HER2-BCH demonstrated favorable tumor targeting and image contrast. [18F]AlF-RESCA-HER2-BCH showed a higher SUVmax in both primary tumor and metastases, and a significantly higher target-to-nontarget ratio in metastases than [18F]AlF-NOTA-HER2-BCH. Moreover, [18F]AlF-RESCA-HER2-BCH had lower renal accumulation (43.56 ± 7.88 vs. 79.81 ± 3.81 at 2 h, p < 0.0001; 33.23 ± 6.89 vs. 78.63 ± 4.00 at 4 h, p < 0.0001) as well as a significantly lower renal absorbed dose than [18F]AlF-NOTA-HER2-BCH (0.4450 ± 0.1117 mGy/MBq vs. 0.8030 ± 0.1604 mGy/MBq, p < 0.01). CONCLUSIONS [18F]AlF-RESCA-HER2-BCH tended to provide better image contrast than [18F]AlF-NOTA-HER2-BCH with a higher target-to-nontarget ratio in detection of metastases. Notably, [18F]AlF-RESCA-HER2-BCH had lower renal accumulation than [18F]AlF-NOTA-HER2-BCH.
Collapse
Affiliation(s)
- Jiayue Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaoyi Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Li Wen
- Guizhou University School of Medicine, Guizhou University, Guiyang, China
| | - Lixin Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Futao Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Guohong Song
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hua Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Nina Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
25
|
Callegari E, Martinelli J, Guidolin N, Boccalon M, Baranyai Z, Tei L. Thermodynamic and Kinetic Stabilities of Al(III) Complexes with N 2O 3 Pentadentate Ligands. Molecules 2023; 28:molecules28093764. [PMID: 37175174 PMCID: PMC10180113 DOI: 10.3390/molecules28093764] [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: 03/20/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Al(III) complexes have been recently investigated for their potential use in imaging with positron emission tomography (PET) by formation of ternary complexes with the radioisotope fluorine-18 (18F). Although the derivatives of 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA) are the most applied chelators for [Al18F]2+ labelling and (pre)clinical PET imaging, non-macrocyclic, semi-rigid pentadentate chelators having two N- and three O-donor atoms such as RESCA1 and AMPDA-HB have been proposed with the aim to allow room temperature labelling of temperature-sensitive biomolecules. The paucity of stability data on Al(III) complexes used for PET imaging instigated a complete thermodynamic and kinetic solution study on Al(III) complexes with aminomethylpiperidine (AMP) derivatives AMPTA and AMPDA-HB and the comparison with a RESCA1-like chelator CD3A-Bn (trans-1,2-diaminocyclohexane-N-benzyl-N,N',N'-triacetic acid). The stability constant of [Al(AMPDA-HB)] is about four orders of magnitude higher than that of [Al(AMPTA)] and [Al(CD3A-Bn)], highlighting the greater affinity of phenolates with respect to acetate O-donors. On the other hand, the kinetic inertness of the complexes, determined by following the Cu2+-mediated transmetallation reactions in the 7.5-10.5 pH range, resulted in a spontaneous and hydroxide-assisted dissociation slightly faster for [Al(AMPTA)] than for the other two complexes (t1/2 = 4.5 h for [Al(AMPTA)], 12.4 h for [Al(AMPDA-HB)], and 24.1 h for [Al(CD3A-Bn)] at pH 7.4 and 25 °C). Finally, the [AlF]2+ ternary complexes were prepared and their stability in reconstituted human serum was determined by 19F NMR experiments.
Collapse
Affiliation(s)
- Edoardo Callegari
- Department of Science and Technological Innovation, Università del Piemonte Orientale, Viale T. Michel 11, 15121 Alessandria, Italy
| | - Jonathan Martinelli
- Department of Science and Technological Innovation, Università del Piemonte Orientale, Viale T. Michel 11, 15121 Alessandria, Italy
| | - Nicol Guidolin
- Bracco Research Centre, Bracco Imaging S.p.A., Via Ribes 5, 10010 Colleretto Giacosa, Italy
| | - Mariangela Boccalon
- Bracco Research Centre, Bracco Imaging S.p.A., Via Ribes 5, 10010 Colleretto Giacosa, Italy
| | - Zsolt Baranyai
- Bracco Research Centre, Bracco Imaging S.p.A., Via Ribes 5, 10010 Colleretto Giacosa, Italy
| | - Lorenzo Tei
- Department of Science and Technological Innovation, Università del Piemonte Orientale, Viale T. Michel 11, 15121 Alessandria, Italy
| |
Collapse
|
26
|
Qin X, Guo X, Liu T, Li L, Zhou N, Ma X, Meng X, Liu J, Zhu H, Jia B, Yang Z. High in-vivo stability in preclinical and first-in-human experiments with [ 18F]AlF-RESCA-MIRC213: a 18F-labeled nanobody as PET radiotracer for diagnosis of HER2-positive cancers. Eur J Nucl Med Mol Imaging 2023; 50:302-313. [PMID: 36129493 DOI: 10.1007/s00259-022-05967-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 09/11/2022] [Indexed: 01/10/2023]
Abstract
PURPOSE [18F]AlF-RESCA was introduced as a core particularly useful for 18F-labeling of heat-sensitive biomolecules. However, no translational studies have been reported up to now. Herein, we reported the first-in-human evaluation of an 18F-labeled anti-HER2 nanobody MIRC213 as a PET radiotracer for imaging HER2-positive cancers. METHODS MIRC213 was produced by E. coli and conjugated with ( ±)-H3RESCA-Mal. [18F]AlF-RESCA-MIRC213 was prepared at room temperature. Its radiochemical purity and stability of were determined by radio-HPLC with the size-exclusion chromatographic column. Cell uptake was performed in NCI-N87 (HER2 +) and MCF-7 (HER2-) cells and the cell-binding affinity was verified in SK-OV-3 (HER2 +) cells. Small-animal PET/CT was performed using SK-OV-3, NCI-N87, and MCF-7 tumor-bearing mice at 30 min, 1 h, and 2 h post-injection. For blocking experiment, excess MIRC213 was co-injected with radiotracer. Biodistribution were performed on SKOV-3 and MCF-7 tumor-bearing mice at 2 h post-injection. For clinical study, PET/CT images were acquired at 2 h and 4 h after injection of [18F]AlF-RESCA-MIRC213 (1.85-3.7 MBq/kg) in six breast cancer patients (3 HER2-positive and 3 HER2-negative). All patients underwent [18F]-FDG PET/CT within a week for tissue selection purpose. Distribution and dosimetry were calculated. Standardized uptake values (SUV) were measured in tumors and normal organs. RESULTS MIRC213 was produced with > 95% purity and modified with RESCA to obtain RESCA-MIRC213. [18F]AlF-RESCA-MIRC213 was prepared within 20 min at room temperature with the radiochemical yield of 50.48 ± 7.6% and radiochemical purity of > 98% (n > 10), and remained stable in both PBS (88%) and 5% HSA (92%) after 6 h. The 2 h cellular uptake of [18F]AlF-RESCA-MIRC213 in NCI-N87 cells was 11.22 ± 0.60 AD%/105 cells. Its binding affinity Kd value was determined to be 1.23 ± 0.58 nM. Small-animal PET/CT with [18F]AlF-RESCA-MIRC213 can clearly differentiate SK-OV-3 and NCI-N87 tumors from MCF-7 tumors and background with a high uptake of 4.73 ± 1.18 ID%/g and substantially reduced to 1.70 ± 0.13 ID%/g for the blocking group (p < 0.05) in SK-OV-3 tumors at 2 h post-injection. No significant bone radioactivity was seen in the tumor-bearing animals. In all six breast cancer patients, there was no adverse reaction during study. The uptake of [18F]AlF-RESCA-MIRC213 was mainly in lacrimal gland, parotid gland, submandibular gland, thyroid gland, gallbladder, kidneys, liver, and intestines. There was no significant bone radioactivity accumulation in cancer patients. [18F]AlF-RESCA-MIRC213 had significantly higher tumor uptake in lesions from HER2-positive patients than that lesions from HER2-negative patients (SUVmax of 3.62 ± 1.56 vs. 1.41 ± 0.41, p = 0.0012) at 2 h post-injection. The kidneys received the highest radiation dose of 2.42 × 10-1 mGy/MBq, and the effective dose was 1.56 × 10-2 mSv/MBq. CONCLUSIONS [18F]AlF-RESCA-MIRC213 could be prepared with high radiolabeling yield under mild conditions. [18F]AlF-RESCA-MIRC213 has relatively high stability both in vitro and in vivo. The results from clinical transformation suggest that [18F]AlF-RESCA-MIRC213 PET/CT is a safe procedure with favorable pharmacokinetics and dosimetry profile, and it is a promising new PET radiotracer for noninvasive diagnosis of HER2-positive cancers.
Collapse
Affiliation(s)
- Xue Qin
- Guizhou University School of Medicine, Guizhou University, Guiyang, 550025, China
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiaoyi Guo
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Tianyu Liu
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Liqiang Li
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Nina Zhou
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), 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
- Guizhou University School of Medicine, Guizhou University, Guiyang, 550025, China
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiangxi Meng
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), 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
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), 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
- Guizhou University School of Medicine, Guizhou University, Guiyang, 550025, China.
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Bing Jia
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| | - Zhi Yang
- Guizhou University School of Medicine, Guizhou University, Guiyang, 550025, China.
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), 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
|
27
|
Aluminum fluoride-18-labelled indocyanine green as a potential PET imaging agent for hepatic function reserve. J Radioanal Nucl Chem 2022. [DOI: 10.1007/s10967-022-08359-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
28
|
Küppers J, Kürpig S, Bundschuh RA, Essler M, Lütje S. Radiolabeling Strategies of Nanobodies for Imaging Applications. Diagnostics (Basel) 2021; 11:1530. [PMID: 34573872 PMCID: PMC8471529 DOI: 10.3390/diagnostics11091530] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/30/2021] [Accepted: 08/20/2021] [Indexed: 02/06/2023] Open
Abstract
Nanobodies are small recombinant antigen-binding fragments derived from camelid heavy-chain only antibodies. Due to their compact structure, pharmacokinetics of nanobodies are favorable compared to full-size antibodies, allowing rapid accumulation to their targets after intravenous administration, while unbound molecules are quickly cleared from the circulation. In consequence, high signal-to-background ratios can be achieved, rendering radiolabeled nanobodies high-potential candidates for imaging applications in oncology, immunology and specific diseases, for instance in the cardiovascular system. In this review, a comprehensive overview of central aspects of nanobody functionalization and radiolabeling strategies is provided.
Collapse
Affiliation(s)
- Jim Küppers
- Department of Nuclear Medicine, University Hospital Bonn, 53127 Bonn, Germany; (S.K.); (R.A.B.); (M.E.); (S.L.)
| | | | | | | | | |
Collapse
|
29
|
Scroggie KR, Perkins MV, Chalker JM. Reaction of [ 18F]Fluoride at Heteroatoms and Metals for Imaging of Peptides and Proteins by Positron Emission Tomography. Front Chem 2021; 9:687678. [PMID: 34249861 PMCID: PMC8262615 DOI: 10.3389/fchem.2021.687678] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
The ability to radiolabel proteins with [18F]fluoride enables the use of positron emission tomography (PET) for the early detection, staging and diagnosis of disease. The direct fluorination of native proteins through C-F bond formation is, however, a difficult task. The aqueous environments required by proteins severely hampers fluorination yields while the dry, organic solvents that promote nucleophilic fluorination can denature proteins. To circumvent these issues, indirect fluorination methods making use of prosthetic groups that are first fluorinated and then conjugated to a protein have become commonplace. But, when it comes to the radiofluorination of proteins, these indirect methods are not always suited to the short half-life of the fluorine-18 radionuclide (110 min). This review explores radiofluorination through bond formation with fluoride at boron, metal complexes, silicon, phosphorus and sulfur. The potential for these techniques to be used for the direct, aqueous radiolabeling of proteins with [18F]fluoride is discussed.
Collapse
Affiliation(s)
| | | | - Justin M. Chalker
- Institute for Nanoscale Science and Technology, College of Science and Engineering, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
30
|
Zha Z, Choi SR, Ploessl K, Alexoff D, Zhao R, Zhu L, Kung HF. Radiolabeling Optimization and Preclinical Evaluation of the New PSMA Imaging Agent [ 18F]AlF-P16-093. Bioconjug Chem 2021; 32:1017-1026. [PMID: 33872489 DOI: 10.1021/acs.bioconjchem.1c00177] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Prostate-specific membrane antigen (PSMA)-targeted radioligands have played an increasing role in the diagnosis of prostate cancer. [68Ga]Ga-P16-093 is a PSMA-targeting agent for positron emission tomography imaging, currently under a Phase 2 clinical trial. In the present study, P16-093 was labeled with 18F via [18F]AlF2+ complex formation, and the biological properties of [18F]AlF-P16-093 were evaluated. Optimization of radiolabeling efficiency was performed by testing a series of parameters, including the amount of free ligand; the amount of Al3+; and the influence of solvent, pH, temperature, reaction time, and reaction volume. Optimal labeling results were achieved at pH 5 by reacting at 60 °C for 15 min in a vial containing 74-370 MBq of [18F]fluoride, 46 nmol of P16-093, 40 nmol of AlCl3·6 H2O, and 50% EtOH. [18F]AlF-P16-093 was prepared with a non-decay-corrected radiochemical yield of 54.4 ± 4.4% (n = 9) within 30 min (final radiochemical purity ≥95%). In vitro, [18F]AlF-P16-093 showed PSMA-specific high uptakes in PIP-PC3 cells. The binding affinity of [18F]AlF-P16-093 to PSMA was determined as Kd of 12.4 ± 2.0 nM. The tumor uptake in mice with a xenografted PSMA-expressing PIP-PC3 tumor was high (18.8 ± 5.14% ID/g at 1 h postinjection) and retained without washout for 2 h. In addition, tumor uptake was almost completely blocked by coinjecting a PSMA inhibitor, 2-PMPA. The bone activity at 1 h post injection was higher with [18F]AlF-P16-093 (2.83 ± 0.49% ID/g) in comparison to that of [68Ga]Ga-P16-093 (0.26 ± 0.07% ID/g). In summary, an efficient and simple radiosynthesis of [18F]AlF-P16-093 was achieved. [18F]AlF-P16-093 showed desirable in vivo pharmacokinetics and excellent PSMA-targeting properties for imaging PSMA expression in prostate cancer.
Collapse
Affiliation(s)
- Zhihao Zha
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
| | - Seok Rye Choi
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
| | - Karl Ploessl
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
| | - David Alexoff
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
| | - Ruiyue Zhao
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Lin Zhu
- College of Chemistry, Beijing Normal University, Beijing 100875, PR China
| | - Hank F Kung
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
31
|
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
|
32
|
Jiang X, Wang X, Shen T, Yao Y, Chen M, Li Z, Li X, Shen J, Kou Y, Chen S, Zhou X, Luo Z, Cheng Z. FAPI-04 PET/CT Using [ 18F]AlF Labeling Strategy: Automatic Synthesis, Quality Control, and In Vivo Assessment in Patient. Front Oncol 2021; 11:649148. [PMID: 33816303 PMCID: PMC8017320 DOI: 10.3389/fonc.2021.649148] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 01/27/2021] [Indexed: 12/20/2022] Open
Abstract
68Ga labeled FAPI is the current standard for FAPI-PET, but its batch activity is limited. [18F]AlF-NOTA-FAPI-04 is a promising alternative combining the advantages of a chelator-based radiolabeling method with the unique properties of fluorine-18. The objective of this study was to develop a quick automatic method for synthesis of [18F]AlF-NOTA-FAPI-04 using a AllinOne synthesis system, and perform PET imaging with [18F]AlF-NOTA-FAPI-04 on patients. [18F]AlF-NOTA-FAPI-04 was produced, and its quality control was conducted by HPLC equipped with a radioactive detector. [18F]AlF-NOTA-FAPI-04 PET/CT imaging was performed in normal BALB/c mice (n = 3) and 4T1 breast cancer models (n = 3) to determine its biodistribution. Then [18F]AlF-NOTA-FAPI-04 and 18F-fluorodeoxyglucose (FDG) PET/CT imaging were performed in an invasive ductal carcinoma patient (female, 54 years old). The synthesis time of [18F]AlF-NOTA-FAPI-04 was about 25 min, and the radiochemical yield was 26.4 ± 1.5% (attenuation correction, n = 10). The radiochemical purity was above 99.0% and was above 98.0% after 6 h. The product was colorless transparent solution with pH value of 7.0-7.5, and the specific activity was 49.41 ± 3.19 GBq/μmol. PET/CT imaging in mice showed that physiological uptake of [18F]AlF-NOTA-FAPI-04 was mainly in the biliary system and bladder, and [18F]AlF-NOTA-FAPI-04 highly concentrated in tumor xenografts. PET/CT imaging in the patient showed that [18F]AlF-NOTA-FAPI-04 obtained high tumor background ratio (TBR) value of 8.44 in segment V and VI, while TBR value was 2.55 by 18F-FDG. [18F]AlF-NOTA-FAPI-04 could be synthesized with high radiochemical yield and batch production by AllinOne module and show excellent diagnosis performance in cancer patients.
Collapse
Affiliation(s)
- Xiao Jiang
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China.,Institute of Isotope, China Institute of Atomic Energy, Beijing, China
| | - Xiaoxiong Wang
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Taipeng Shen
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Yutang Yao
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Meihua Chen
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Zeng Li
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Xiuli Li
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Jiaqi Shen
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Ying Kou
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Shirong Chen
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Xing Zhou
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Zhifu Luo
- Institute of Isotope, China Institute of Atomic Energy, Beijing, China
| | - Zhuzhong Cheng
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| |
Collapse
|
33
|
Piramoon M, Khodadust F, Hosseinimehr SJ. Radiolabeled nanobodies for tumor targeting: From bioengineering to imaging and therapy. Biochim Biophys Acta Rev Cancer 2021; 1875:188529. [PMID: 33647388 DOI: 10.1016/j.bbcan.2021.188529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 02/08/2023]
Abstract
So far, numerous molecules and biomolecules have been evaluated for tumor targeting purposes for radionuclide-based imaging and therapy modalities. Due to the high affinity and specificity against tumor antigens, monoclonal antibodies are appropriate candidates for tumor targeting. However, their large size prevents their comprehensive application in radionuclide-based tumor imaging or therapy, since it leads to their low tumor penetration, low blood clearance, and thus inappropriate tumor-to-background ratio. Nowadays, the variable domain of heavy-chain antibodies from the Camelidae family, known as nanobodies (Nbs), turn into exciting candidates for medical research. Considering several innate advantages of these new tumor-targeting agents, including excellent affinity and specificity toward antigen, high solubility, high stability, fast washout from blood, convenient production, ease of selection, and low immunogenicity, it assumes that they may overcome generic problems of monoclonal antibodies, their fragments, and other vectors used for tumor imaging/therapy. After three decades of Nbs discovery, the increasing number of their preclinical and clinical investigations, which have led to outstanding results, confirm their application for tumor targeting purposes. This review describes Nbs characteristics, the diagnostic and therapeutic application of their radioconjugates, and their recent advances.
Collapse
Affiliation(s)
- Majid Piramoon
- Department of Medicinal Chemistry and Radiopharmacy, School of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran; Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Fatemeh Khodadust
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran; Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
34
|
Ranjbar Bahadori S, Mulgaonkar A, Hart R, Wu CY, Zhang D, Pillai A, Hao Y, Sun X. Radiolabeling strategies and pharmacokinetic studies for metal based nanotheranostics. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 13:e1671. [PMID: 33047504 DOI: 10.1002/wnan.1671] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 12/17/2022]
Abstract
Radiolabeled metal-based nanoparticles (MNPs) have drawn considerable attention in the fields of nuclear medicine and molecular imaging, drug delivery, and radiation therapy, given the fact that they can be potentially used as diagnostic imaging and/or therapeutic agents, or even as theranostic combinations. Here, we present a systematic review on recent advances in the design and synthesis of MNPs with major focuses on their radiolabeling strategies and the determinants of their in vivo pharmacokinetics, and together how their intended applications would be impacted. For clarification, we categorize all reported radiolabeling strategies for MNPs into indirect and direct approaches. While indirect labeling simply refers to the use of bifunctional chelators or prosthetic groups conjugated to MNPs for post-synthesis labeling with radionuclides, we found that many practical direct labeling methodologies have been developed to incorporate radionuclides into the MNP core without using extra reagents, including chemisorption, radiochemical doping, hadronic bombardment, encapsulation, and isotope or cation exchange. From the perspective of practical use, a few relevant examples are presented and discussed in terms of their pros and cons. We further reviewed the determinants of in vivo pharmacokinetic parameters of MNPs, including factors influencing their in vivo absorption, distribution, metabolism, and elimination, and discussed the challenges and opportunities in the development of radiolabeled MNPs for in vivo biomedical applications. Taken together, we believe the cumulative advancement summarized in this review would provide a general guidance in the field for design and synthesis of radiolabeled MNPs towards practical realization of their much desired theranostic capabilities. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Diagnostic Tools > Diagnostic Nanodevices Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Shahab Ranjbar Bahadori
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Aditi Mulgaonkar
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ryan Hart
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Cheng-Yang Wu
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Dianbo Zhang
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Anil Pillai
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yaowu Hao
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Xiankai Sun
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
35
|
Lin D, Wallace M, Allentoff AJ, Donnelly DJ, Gomes E, Voronin K, Gong S, Huang RYC, Kim H, Caceres-Cortes J, Bonacorsi S. Chemoselective Methionine Bioconjugation: Site-Selective Fluorine-18 Labeling of Proteins and Peptides. Bioconjug Chem 2020; 31:1908-1916. [PMID: 32687313 DOI: 10.1021/acs.bioconjchem.0c00256] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
36
|
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
|
37
|
Bridoux J, Neyt S, Debie P, Descamps B, Devoogdt N, Cleeren F, Bormans G, Broisat A, Caveliers V, Xavier C, Vanhove C, Hernot S. Improved Detection of Molecular Markers of Atherosclerotic Plaques Using Sub-Millimeter PET Imaging. Molecules 2020; 25:molecules25081838. [PMID: 32316285 PMCID: PMC7221983 DOI: 10.3390/molecules25081838] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/13/2020] [Accepted: 04/13/2020] [Indexed: 12/14/2022] Open
Abstract
Since atherosclerotic plaques are small and sparse, their non-invasive detection via PET imaging requires both highly specific radiotracers as well as imaging systems with high sensitivity and resolution. This study aimed to assess the targeting and biodistribution of a novel fluorine-18 anti-VCAM-1 Nanobody (Nb), and to investigate whether sub-millimetre resolution PET imaging could improve detectability of plaques in mice. The anti-VCAM-1 Nb functionalised with the novel restrained complexing agent (RESCA) chelator was labelled with [18F]AlF with a high radiochemical yield (>75%) and radiochemical purity (>99%). Subsequently, [18F]AlF(RESCA)-cAbVCAM1-5 was injected in ApoE-/- mice, or co-injected with excess of unlabelled Nb (control group). Mice were imaged sequentially using a cross-over design on two different commercially available PET/CT systems and finally sacrificed for ex vivo analysis. Both the PET/CT images and ex vivo data showed specific uptake of [18F]AlF(RESCA)-cAbVCAM1-5 in atherosclerotic lesions. Non-specific bone uptake was also noticeable, most probably due to in vivo defluorination. Image analysis yielded higher target-to-heart and target-to-brain ratios with the β-CUBE (MOLECUBES) PET scanner, demonstrating that preclinical detection of atherosclerotic lesions could be improved using the latest PET technology.
Collapse
Affiliation(s)
- Jessica Bridoux
- Laboratory of In Vivo Cellular and Molecular Imaging (ICMI, BEFY-MIMA), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; (J.B.); (P.D.); (N.D.); (V.C.); (C.X.)
| | - Sara Neyt
- Preclinical imaging, MOLECUBES NV, 9000 Ghent, Belgium;
| | - Pieterjan Debie
- Laboratory of In Vivo Cellular and Molecular Imaging (ICMI, BEFY-MIMA), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; (J.B.); (P.D.); (N.D.); (V.C.); (C.X.)
| | | | - Nick Devoogdt
- Laboratory of In Vivo Cellular and Molecular Imaging (ICMI, BEFY-MIMA), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; (J.B.); (P.D.); (N.D.); (V.C.); (C.X.)
| | - Frederik Cleeren
- Radiopharmaceutical Research, KU Leuven, 3000 Leuven, Belgium; (F.C.); (G.B.)
| | - Guy Bormans
- Radiopharmaceutical Research, KU Leuven, 3000 Leuven, Belgium; (F.C.); (G.B.)
| | - Alexis Broisat
- Radiopharmaceutiques Biocliniques, INSERM 1039, Université de Grenoble, 38400 Grenoble, France;
| | - Vicky Caveliers
- Laboratory of In Vivo Cellular and Molecular Imaging (ICMI, BEFY-MIMA), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; (J.B.); (P.D.); (N.D.); (V.C.); (C.X.)
- Nuclear Medicine department, UZ Brussel, 1090 Brussels, Belgium
| | - Catarina Xavier
- Laboratory of In Vivo Cellular and Molecular Imaging (ICMI, BEFY-MIMA), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; (J.B.); (P.D.); (N.D.); (V.C.); (C.X.)
| | - Christian Vanhove
- IBiTech-MEDISIP, Ghent University, 9000 Ghent, Belgium; (B.D.); (C.V.)
| | - Sophie Hernot
- Laboratory of In Vivo Cellular and Molecular Imaging (ICMI, BEFY-MIMA), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; (J.B.); (P.D.); (N.D.); (V.C.); (C.X.)
- Correspondence: ; Tel.: +32-2-477-49-91
| |
Collapse
|
38
|
van der Veen EL, Suurs FV, Cleeren F, Bormans G, Elsinga PH, Hospers GAP, Lub-de Hooge MN, de Vries EGE, de Vries EFJ, Antunes IF. Development and Evaluation of Interleukin-2-Derived Radiotracers for PET Imaging of T Cells in Mice. J Nucl Med 2020; 61:1355-1360. [PMID: 32111688 DOI: 10.2967/jnumed.119.238782] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/03/2020] [Indexed: 12/14/2022] Open
Abstract
Recently, N-(4-18F-fluorobenzoyl)-interleukin-2 (18F-FB-IL2) was introduced as a PET tracer for T cell imaging. However, production is complex and time-consuming. Therefore, we developed 2 radiolabeled IL2 variants, namely aluminum 18F-fluoride-(restrained complexing agent)-IL2 (18F-AlF-RESCA-IL2) and 68Ga-gallium-(1,4,7-triazacyclononane-4,7-diacetic acid-1-glutaric acid)-IL2 (68Ga-Ga-NODAGA-IL2), and compared their in vitro and in vivo characteristics with 18F-FB-IL2. Methods: Radiolabeling of 18F-AlF-RESCA-IL2 and 68Ga-Ga-NODAGA-IL2 was optimized, and stability was evaluated in human serum. Receptor binding was studied with activated human peripheral blood mononuclear cells (hPBMCs). Ex vivo tracer biodistribution in immunocompetent BALB/cOlaHsd (BALB/c) mice was performed at 15, 60, and 90 min after tracer injection. In vivo binding characteristics were studied in severe combined immunodeficient (SCID) mice inoculated with activated hPBMCs in Matrigel. Tracer was injected 15 min after hPBMC inoculation, and a 60-min dynamic PET scan was acquired, followed by ex vivo biodistribution studies. Specific uptake was determined by coinjection of tracer with unlabeled IL2 and by evaluating uptake in a control group inoculated with Matrigel only. Results: 68Ga-Ga-NODAGA-IL2 and 18F-AlF-RESCA-IL2 were produced with radiochemical purity of more than 95% and radiochemical yield of 13.1% ± 4.7% and 2.4% ± 1.6% within 60 and 90 min, respectively. Both tracers were stable in serum, with more than 90% being intact tracer after 1 h. In vitro, both tracers displayed preferential binding to activated hPBMCs. Ex vivo biodistribution studies on BALB/c mice showed higher uptake of 18F-AlF-RESCA-IL2 than of 18F-FB-IL2 in liver, kidney, spleen, bone, and bone marrow. 68Ga-Ga-NODAGA-IL2 uptake in liver and kidney was higher than 18F-FB-IL2 uptake. In vivo, all tracers revealed uptake in activated hPBMCs in SCID mice. Low uptake was seen after a blocking dose of IL2 and in the Matrigel control group. In addition, 18F-AlF-RESCA-IL2 yielded the highest-contrast PET images of target lymph nodes. Conclusion: Production of 18F-AlF-RESCA-IL2 and 68Ga-Ga-NODAGA-IL2 is simpler and faster than that of 18F-FB-IL2. Both tracers showed good in vitro and in vivo characteristics, with high uptake in lymphoid tissue and hPBMC xenografts.
Collapse
Affiliation(s)
- Elly L van der Veen
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Frans V Suurs
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Frederik Cleeren
- Laboratory for Radiopharmaceutical Research, Department of Pharmacy and Pharmacology, University of Leuven, Leuven, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmaceutical Research, Department of Pharmacy and Pharmacology, University of Leuven, Leuven, Belgium
| | - Philip H Elsinga
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and
| | - Geke A P Hospers
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marjolijn N Lub-de Hooge
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and.,Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Erik F J de Vries
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and
| | - Inês F Antunes
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and
| |
Collapse
|
39
|
Tshibangu T, Cawthorne C, Serdons K, Pauwels E, Gsell W, Bormans G, Deroose CM, Cleeren F. Automated GMP compliant production of [ 18F]AlF-NOTA-octreotide. EJNMMI Radiopharm Chem 2020; 5:4. [PMID: 31997090 PMCID: PMC6989705 DOI: 10.1186/s41181-019-0084-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/16/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Gallium-68 labeled synthetic somatostatin analogs for PET/CT imaging are the current gold standard for somatostatin receptor imaging in neuroendocrine tumor patients. Despite good imaging properties, their use in clinical practice is hampered by the low production levels of 68Ga eluted from a 68Ge/68Ga generator. In contrast, 18F-tracers can be produced in large quantities allowing centralized production and distribution to distant PET centers. [18F]AlF-NOTA-octreotide is a promising tracer that combines a straightforward Al18F-based production procedure with excellent in vivo pharmacokinetics and specific tumor uptake, demonstrated in SSTR2 positive tumor mice. However, advancing towards clinical studies with [18F]AlF-NOTA-octreotide requires the development of an efficient automated GMP production process and additional preclinical studies are necessary to further evaluate the in vivo properties of [18F]AlF-NOTA-octreotide. In this study, we present the automated GMP production of [18F]AlF-NOTA-octreotide on the Trasis AllinOne® radio-synthesizer platform and quality control of the drug product in accordance with GMP. Further, radiometabolite studies were performed and the pharmacokinetics and biodistribution of [18F]AlF-NOTA-octreotide were assessed in healthy rats using μPET/MR. RESULTS The production process of [18F]AlF-NOTA-octreotide has been validated by three validation production runs and the tracer was obtained with a final batch activity of 10.8 ± 1.3 GBq at end of synthesis with a radiochemical yield of 26.1 ± 3.6% (dc), high radiochemical purity and stability (96.3 ± 0.2% up to 6 h post synthesis) and an apparent molar activity of 160.5 ± 75.3 GBq/μmol. The total synthesis time was 40 ± 3 min. Further, the quality control was successfully implemented using validated analytical procedures. Finally, [18F]AlF-NOTA-octreotide showed high in vivo stability and favorable pharmacokinetics with high and specific accumulation in SSTR2-expressing organs in rats. CONCLUSION This robust and automated production process provides high batch activity of [18F]AlF-NOTA-octreotide allowing centralized production and shipment of the compound to remote PET centers. Further, the production process and quality control developed for [18F]AlF-NOTA-octreotide is easily implementable in a clinical setting and the tracer is a potential clinical alternative for somatostatin directed 68Ga labeled peptides obviating the need for a 68Ge/68Ga-generator. Finally, the favorable in vivo properties of [18F]AlF-NOTA-octreotide in rats, with high and specific accumulation in SSTR2 expressing organs, supports clinical translation.
Collapse
Affiliation(s)
- Térence Tshibangu
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Herestraat 49 Box 821, 3000 Leuven, Belgium
| | - Christopher Cawthorne
- Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Biomedical MRI/MoSAIC, Department of Imaging and Pathology, Biomedical Sciences Group, KU Leuven, Leuven, Belgium
| | - Kim Serdons
- Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Elin Pauwels
- Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Willy Gsell
- Biomedical MRI/MoSAIC, Department of Imaging and Pathology, Biomedical Sciences Group, KU Leuven, Leuven, Belgium
| | - Guy Bormans
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Herestraat 49 Box 821, 3000 Leuven, Belgium
| | - Christophe M. Deroose
- Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Frederik Cleeren
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Herestraat 49 Box 821, 3000 Leuven, Belgium
| |
Collapse
|
40
|
Kee C, Tack O, Guibbal F, Wilson TC, Isenegger PG, Imiołek M, Verhoog S, Tilby M, Boscutti G, Ashworth S, Chupin J, Kashani R, Poh AWJ, Sosabowski JK, Macholl S, Plisson C, Cornelissen B, Willis MC, Passchier J, Davis BG, Gouverneur V. 18F-Trifluoromethanesulfinate Enables Direct C-H 18F-Trifluoromethylation of Native Aromatic Residues in Peptides. J Am Chem Soc 2020; 142:1180-1185. [PMID: 31913613 PMCID: PMC6978814 DOI: 10.1021/jacs.9b11709] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Indexed: 12/27/2022]
Abstract
18F labeling strategies for unmodified peptides with [18F]fluoride require 18F-labeled prosthetics for bioconjugation more often with cysteine thiols or lysine amines. Here we explore selective radical chemistry to target aromatic residues applying C-H 18F-trifluoromethylation. We report a one-step route to [18F]CF3SO2NH4 from [18F]fluoride and its application to direct [18F]CF3 incorporation at tryptophan or tyrosine residues using unmodified peptides as complex as recombinant human insulin. The fully automated radiosynthesis of octreotide[Trp(2-CF218F)] enables in vivo positron emission tomography imaging.
Collapse
Affiliation(s)
- Choon
Wee Kee
- Chemistry
Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K.
| | - Osman Tack
- Chemistry
Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K.
| | - Florian Guibbal
- Chemistry
Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K.
- Radiobiology
Research Institute, Department of Oncology, University of Oxford, Headington, Oxford OX3 7LJ, U.K.
| | - Thomas C. Wilson
- Chemistry
Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K.
| | - Patrick G. Isenegger
- Chemistry
Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K.
| | - Mateusz Imiołek
- Chemistry
Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K.
| | - Stefan Verhoog
- Chemistry
Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K.
| | - Michael Tilby
- Chemistry
Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K.
| | | | | | - Juliette Chupin
- Invicro
Ltd, Du Cane Road, London W12 0NN, U.K.
- Centre
for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, U.K.
| | - Roxana Kashani
- Centre
for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, U.K.
| | - Adeline W. J. Poh
- Chemistry
Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K.
| | - Jane K. Sosabowski
- Centre
for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, U.K.
| | - Sven Macholl
- Invicro
Ltd, Du Cane Road, London W12 0NN, U.K.
- Centre
for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, U.K.
| | | | - Bart Cornelissen
- Radiobiology
Research Institute, Department of Oncology, University of Oxford, Headington, Oxford OX3 7LJ, U.K.
| | - Michael C. Willis
- Chemistry
Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K.
| | | | - Benjamin G. Davis
- Chemistry
Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K.
| | - Véronique Gouverneur
- Chemistry
Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K.
| |
Collapse
|
41
|
Russelli L, Martinelli J, De Rose F, Reder S, Herz M, Schwaiger M, Weber W, Tei L, D'Alessandria C. Room Temperature Al 18 F Labeling of 2-Aminomethylpiperidine-Based Chelators for PET Imaging. ChemMedChem 2020; 15:284-292. [PMID: 31830368 DOI: 10.1002/cmdc.201900652] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/02/2019] [Indexed: 01/14/2023]
Abstract
Positron emission tomography (PET) is a non-invasive molecular imaging technology that is constantly expanding, with a high demand for specific antibody-derived imaging probes. The use of tracers based on temperature-sensitive molecules (i. e. Fab, svFab, nanobodies) is increasing and has led us to design a class of chelators based on the structure of 2-aminomethylpiperidine (AMP) with acetic and/or hydroxybenzyl pendant arms (2-AMPTA, NHB-2-AMPDA, and 2-AMPDA-HB), which were investigated as such for {Al18 F}2+ -core chelation efficiency. All the compounds were characterized by HPLC-MS analysis and NMR spectroscopy. The AlF-18 labeling reactions were performed under various conditions (pH/temperature), and the radiolabeled chelates were purified and characterized by radio-TLC and radio-HPLC. The stability of labeled chelates was investigated up to 240 min in human serum (HS), EDTA 5 mM, PBS and 0.9 % NaCl solutions. The in vivo stability of [Al18 F(2-AMPDA-HB)]- was assessed in healthy nude mice (n=6). Radiochemical yields between 55 % and 81 % were obtained at pH 5 and room temperature. High stability in HS was measured for [Al18 F(2-AMPDA-HB)]- , with 90 % of F-18 complexed after 120 min. High stability in vivo, rapid hepatobiliary and renal excretion, with low accumulation of free F-18 in bones were measured. Thus, this new Al18 F-chelator may have a great impact on immuno-PET radiopharmacy, by facilitating the development of new fluorine-18-labeled heat-sensitive biomolecules.
Collapse
Affiliation(s)
- Lisa Russelli
- Department of Nuclear Medicine, Klinikum rechts der Isar TU München, Ismaningerstraße 22, 81675, Munich, Germany
| | - Jonathan Martinelli
- Department of Science and Technological Innovation, Università del Piemonte Orientale, Viale T. Michel 11, 15121, Alessandria, Italy
| | - Francesco De Rose
- Department of Nuclear Medicine, Klinikum rechts der Isar TU München, Ismaningerstraße 22, 81675, Munich, Germany
| | - Sybille Reder
- Department of Nuclear Medicine, Klinikum rechts der Isar TU München, Ismaningerstraße 22, 81675, Munich, Germany
| | - Michael Herz
- Department of Nuclear Medicine, Klinikum rechts der Isar TU München, Ismaningerstraße 22, 81675, Munich, Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine, Klinikum rechts der Isar TU München, Ismaningerstraße 22, 81675, Munich, Germany
| | - Wolfgang Weber
- Department of Nuclear Medicine, Klinikum rechts der Isar TU München, Ismaningerstraße 22, 81675, Munich, Germany
| | - Lorenzo Tei
- Department of Science and Technological Innovation, Università del Piemonte Orientale, Viale T. Michel 11, 15121, Alessandria, Italy
| | - Calogero D'Alessandria
- Department of Nuclear Medicine, Klinikum rechts der Isar TU München, Ismaningerstraße 22, 81675, Munich, Germany
| |
Collapse
|
42
|
Klenner MA, Zhang B, Ciancaleoni G, Howard JK, Maynard-Casely HE, Clegg JK, Massi M, Fraser BH, Pascali G. Rhenium(i) complexation–dissociation strategy for synthesising fluorine-18 labelled pyridine bidentate radiotracers. RSC Adv 2020; 10:8853-8865. [PMID: 35496512 PMCID: PMC9049978 DOI: 10.1039/d0ra00318b] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 02/10/2020] [Indexed: 01/23/2023] Open
Abstract
A novel fluorine-18 radiolabelling method employing rhenium(i) mediation is described herein. In less than 1 minute, fluorine-18 labelled complexes and ligands were synthesised in greater than 80% and 60% radiochemical yields (RCY), respectively.
Collapse
Affiliation(s)
- Mitchell A. Klenner
- Human Health & National Deuteration Facility
- Australian Nuclear Science and Technology Organisation (ANSTO)
- Australia
- School of Molecular and Life Sciences
- Curtin University
| | - Bo Zhang
- School of Chemistry
- Monash University
- Melbourne
- Australia
| | | | - James K. Howard
- Human Health & National Deuteration Facility
- Australian Nuclear Science and Technology Organisation (ANSTO)
- Australia
| | - Helen E. Maynard-Casely
- Human Health & National Deuteration Facility
- Australian Nuclear Science and Technology Organisation (ANSTO)
- Australia
| | - Jack K. Clegg
- School of Chemistry and Molecular Biosciences
- The University of Queensland
- St. Lucia
- Australia
| | | | - Benjamin H. Fraser
- Human Health & National Deuteration Facility
- Australian Nuclear Science and Technology Organisation (ANSTO)
- Australia
| | - Giancarlo Pascali
- Human Health & National Deuteration Facility
- Australian Nuclear Science and Technology Organisation (ANSTO)
- Australia
- Brain and Mind Centre
- The University of Sydney
| |
Collapse
|
43
|
Fersing C, Bouhlel A, Cantelli C, Garrigue P, Lisowski V, Guillet B. A Comprehensive Review of Non-Covalent Radiofluorination Approaches Using Aluminum [ 18F]fluoride: Will [ 18F]AlF Replace 68Ga for Metal Chelate Labeling? Molecules 2019; 24:E2866. [PMID: 31394799 PMCID: PMC6719958 DOI: 10.3390/molecules24162866] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 07/31/2019] [Accepted: 08/06/2019] [Indexed: 12/20/2022] Open
Abstract
Due to its ideal physical properties, fluorine-18 turns out to be a key radionuclide for positron emission tomography (PET) imaging, for both preclinical and clinical applications. However, usual biomolecules radiofluorination procedures require the formation of covalent bonds with fluorinated prosthetic groups. This drawback makes radiofluorination impractical for routine radiolabeling, gallium-68 appearing to be much more convenient for the labeling of chelator-bearing PET probes. In response to this limitation, a recent expansion of the 18F chemical toolbox gave aluminum [18F]fluoride chemistry a real prominence since the late 2000s. This approach is based on the formation of an [18F][AlF]2+ cation, complexed with a 9-membered cyclic chelator such as NOTA, NODA or their analogs. Allowing a one-step radiofluorination in an aqueous medium, this technique combines fluorine-18 and non-covalent radiolabeling with the advantage of being very easy to implement. Since its first reports, [18F]AlF radiolabeling approach has been applied to a wide variety of potential PET imaging vectors, whether of peptidic, proteic, or small molecule structure. Most of these [18F]AlF-labeled tracers showed promising preclinical results and have reached the clinical evaluation stage for some of them. The aim of this report is to provide a comprehensive overview of [18F]AlF labeling applications through a description of the various [18F]AlF-labeled conjugates, from their radiosynthesis to their evaluation as PET imaging agents.
Collapse
Affiliation(s)
- Cyril Fersing
- Institut de Recherche en Cancérologie de Montpellier (IRCM), University of Montpellier, INSERM U1194, Montpellier Cancer Institute (ICM), 34298 Montpellier, France.
- Nuclear Medicine Department, Montpellier Cancer Institute (ICM), University of Montpellier, 208 Avenue des Apothicaires, 34298 Montpellier CEDEX 5, France.
| | - Ahlem Bouhlel
- CERIMED, Aix-Marseille University, 13005 Marseille, France
- Centre de recherche en CardioVasculaire et Nutrition (C2VN), Aix-Marseille University, INSERM 1263, INRA 1260, 13385 Marseille, France
| | - Christophe Cantelli
- Institut de Recherche en Cancérologie de Montpellier (IRCM), University of Montpellier, INSERM U1194, Montpellier Cancer Institute (ICM), 34298 Montpellier, France
- Institut des Biomolécules Max Mousseron, UMR 5247, CNRS, Université de Montpellier, ENSCM, UFR des Sciences Pharmaceutiques et Biologiques, 34093 Montpellier CEDEX, France
| | - Philippe Garrigue
- CERIMED, Aix-Marseille University, 13005 Marseille, France
- Centre de recherche en CardioVasculaire et Nutrition (C2VN), Aix-Marseille University, INSERM 1263, INRA 1260, 13385 Marseille, France
- Department of Nuclear Medicine, Aix-Marseille University, Assistance Publique-Hôpitaux de Marseille (AP-HM), 13385 Marseille, France
| | - Vincent Lisowski
- Institut des Biomolécules Max Mousseron, UMR 5247, CNRS, Université de Montpellier, ENSCM, UFR des Sciences Pharmaceutiques et Biologiques, 34093 Montpellier CEDEX, France
| | - Benjamin Guillet
- CERIMED, Aix-Marseille University, 13005 Marseille, France
- Centre de recherche en CardioVasculaire et Nutrition (C2VN), Aix-Marseille University, INSERM 1263, INRA 1260, 13385 Marseille, France
- Department of Nuclear Medicine, Aix-Marseille University, Assistance Publique-Hôpitaux de Marseille (AP-HM), 13385 Marseille, France
| |
Collapse
|
44
|
Vermeulen K, Vandamme M, Bormans G, Cleeren F. Design and Challenges of Radiopharmaceuticals. Semin Nucl Med 2019; 49:339-356. [PMID: 31470930 DOI: 10.1053/j.semnuclmed.2019.07.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
This review describes general concepts with regard to radiopharmaceuticals for diagnostic or therapeutic applications that help to understand the specific challenges encountered during the design, (radio)synthesis, in vitro and in vivo evaluation and clinical translation of novel radiopharmaceuticals. The design of a radiopharmaceutical requires upfront decisions with regard to combining a suitable vector molecule with an appropriate radionuclide, considering the type and location of the molecular target, the desired application, and the time constraints imposed by the relatively short half-life of radionuclides. Well-designed in vitro and in vivo experiments allow nonclinical validation of radiotracers. Ultimately, in combination with a limited toxicology package, the radiotracer becomes a radiopharmaceutical for clinical evaluation, produced in compliance with regulatory requirements for medicines for intravenous (IV) injection.
Collapse
Affiliation(s)
- Koen Vermeulen
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| | - Mathilde Vandamme
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium.
| | - Frederik Cleeren
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| |
Collapse
|
45
|
NIRF-Molecular Imaging with Synovial Macrophages-Targeting Vsig4 Nanobody for Disease Monitoring in a Mouse Model of Arthritis. Int J Mol Sci 2019; 20:ijms20133347. [PMID: 31288389 PMCID: PMC6651725 DOI: 10.3390/ijms20133347] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/05/2019] [Accepted: 07/06/2019] [Indexed: 01/21/2023] Open
Abstract
Nanobody against V-set and Ig domain-containing 4 (Vsig4) on tissue macrophages, such as synovial macrophages, could visualize joint inflammation in multiple experimental arthritis models via single-photon emission computed tomography imaging. Here, we further addressed the specificity and assessed the potential for arthritis monitoring using near-infrared fluorescence (NIRF) Cy7-labeled Vsig4 nanobody (Cy7-Nb119). In vivo NIRF-imaging of collagen-induced arthritis (CIA) was performed using Cy7-Nb119. Signals obtained with Cy7-Nb119 or isotope control Cy7-NbBCII10 were compared in joints of naive mice versus CIA mice. In addition, pathological microscopy and fluorescence microscopy were used to validate the arthritis development in CIA. Cy7-Nb119 accumulated in inflamed joints of CIA mice, but not the naive mice. Development of symptoms in CIA was reflected in increased joint accumulation of Cy7-Nb119, which correlated with the conventional measurements of disease. Vsig4 is co-expressed with F4/80, indicating targeting of the increasing number of synovial macrophages associated with the severity of inflammation by the Vsig4 nanobody. NIRF imaging with Cy7-Nb119 allows specific assessment of inflammation in experimental arthritis and provides complementary information to clinical scoring for quantitative, non-invasive and economical monitoring of the pathological process. Nanobody labelled with fluorescence can also be used for ex vivo validation experiments using flow cytometry and fluorescence microscopy.
Collapse
|
46
|
Zhou Z, Devoogdt N, Zalutsky MR, Vaidyanathan G. An Efficient Method for Labeling Single Domain Antibody Fragments with 18F Using Tetrazine- Trans-Cyclooctene Ligation and a Renal Brush Border Enzyme-Cleavable Linker. Bioconjug Chem 2018; 29:4090-4103. [PMID: 30384599 DOI: 10.1021/acs.bioconjchem.8b00699] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Single domain antibody fragments (sdAbs) labeled with 18F have shown promise for assessing the status of oncological targets such as the human epidermal growth factor receptor 2 (HER2) by positron emission tomography (PET). Earlier, we evaluated two residualizing prosthetic agents for 18F-labeling of anti-HER2 sdAbs; however, these methods resulted in poor labeling yields and high uptake of 18F activity in the kidneys. To potentially mitigate these limitations, we have now developed an 18F labeling method that utilizes the trans-cyclooctene (TCO)-tetrazine (Tz)-based inverse-electron demand Diels-Alder reaction (IEDDAR) in tandem with a renal brush border enzyme-cleavable glycine-lysine (GK) linker in the prosthetic moiety. The HER2-targeted sdAb 2Rs15d was derivatized with TCO-GK-PEG4-NHS or TCO-PEG4-NHS, which lacks the cleavable linker. As an additional control, the non HER2-specific sdAb R3B23 was derivatized with TCO-GK-PEG4-NHS. The resultant sdAb conjugates were labeled with 18F by IEDDAR using [18F]AlF-NOTA-PEG4-methyltetrazine. As a positive control, the 2Rs15d sdAb was radioiodinated using the well-characterized residualizing prosthetic agent, N-succinimidyl 4-guanidinomethyl-3-[125I]iodobenzoate ([125I]SGMIB). Synthesis of [18F]AlF-NOTA-Tz-TCO-GK-2Rs15d was achieved with an overall radiochemical yield (RCY) of 17.8 ± 1.5% ( n = 5) in 90 min, a significant improvement over prior methods (3-4% in 2-3 h). In vitro assays indicated that [18F]AlF-NOTA-Tz-TCO-GK-2Rs15d bound with high affinity and immunoreactivity to HER2. In normal mice, when normalized to coinjected [125I]SGMIB-2Rs15d, the kidney uptake of [18F]AlF-NOTA-Tz-TCO-GK-2Rs15d was 15- and 28-fold lower ( P < 0.001) than that seen for the noncleavable control ([18F]AlF-NOTA-Tz-TCO-2Rs15d) at 1 and 3 h, respectively. Uptake of [18F]AlF-NOTA-Tz-TCO-GK-2Rs15d in HER2-expressing SKOV-3 ovarian carcinoma xenografts implanted in athymic mice was about 80% of that seen for coinjected [125I]SGMIB-2Rs15d. On the other hand, kidney uptake was 5-6-fold lower, and as a result, tumor-to-kidney ratios were 4-fold higher for [18F]AlF-NOTA-Tz-TCO-GK-2Rs15d than those for [125I]SGMIB-2Rs15d. SKOV-3 xenografts were clearly delineated even at 1 h after administration of [18F]AlF-NOTA-Tz-TCO-GK-2Rs15d by Micro-PET/CT imaging with even higher contrast observed thereafter. In conclusion, this strategy warrants further evaluation for labeling small proteins such as sdAbs because it offers the benefits of good radiochemical yields and enhanced tumor-to-normal tissue ratios, particularly in the kidney.
Collapse
Affiliation(s)
- Zhengyuan Zhou
- Department of Radiology , Duke University Medical Center , Durham , North Carolina 27710 , United States
| | - Nick Devoogdt
- In vivo Cellular and Molecular Imaging laboratory , Vrije Universiteit Brussel , 1090 , Brussels , Belgium
| | - Michael R Zalutsky
- Department of Radiology , Duke University Medical Center , Durham , North Carolina 27710 , United States
| | - Ganesan Vaidyanathan
- Department of Radiology , Duke University Medical Center , Durham , North Carolina 27710 , United States
| |
Collapse
|