1
|
Ebner R, Lohse A, Fabritius MP, Rübenthaler J, Wängler C, Wängler B, Schirrmacher R, Völter F, Schmid HP, Unterrainer LM, Öcal O, Hinterberger A, Spitzweg C, Auernhammer CJ, Geyer T, Ricke J, Bartenstein P, Holzgreve A, Grawe F. Validation of the standardization framework SSTR-RADS 1.0 for neuroendocrine tumors using the novel SSTR‑targeting peptide [ 18F]SiTATE. Eur Radiol 2024; 34:7222-7232. [PMID: 38769164 PMCID: PMC11519286 DOI: 10.1007/s00330-024-10788-3] [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: 11/27/2023] [Revised: 02/27/2024] [Accepted: 04/18/2024] [Indexed: 05/22/2024]
Abstract
OBJECTIVES Somatostatin receptor positron emission tomography/computed tomography (SSTR-PET/CT) using [68Ga]-labeled tracers is a widely used imaging modality for neuroendocrine tumors (NET). Recently, [18F]SiTATE, a SiFAlin tagged [Tyr3]-octreotate (TATE) PET tracer, has shown great potential due to favorable clinical characteristics. We aimed to evaluate the reproducibility of Somatostatin Receptor-Reporting and Data System 1.0 (SSTR-RADS 1.0) for structured interpretation and treatment planning of NET using [18F]SiTATE. METHODS Four readers assessed [18F]SiTATE-PET/CT of 95 patients according to the SSTR-RADS 1.0 criteria at two different time points. Each reader evaluated up to five target lesions per scan. The overall scan score and the decision on peptide receptor radionuclide therapy (PRRT) were considered. Inter- and intra-reader agreement was determined using the intraclass correlation coefficient (ICC). RESULTS The ICC analysis on the inter-reader agreement using SSTR-RADS 1.0 for identical target lesions (ICC ≥ 85%), overall scan score (ICC ≥ 90%), and the decision to recommend PRRT (ICC ≥ 85%) showed excellent agreement. However, significant differences were observed in recommending PRRT among experienced readers (ER) (p = 0.020) and inexperienced readers (IR) (p = 0.004). Compartment-based analysis demonstrated good to excellent inter-reader agreement for most organs (ICC ≥ 74%), except for lymph nodes (ICC ≥ 53%). CONCLUSION SSTR-RADS 1.0 represents a highly reproducible and consistent framework system for stratifying SSTR-targeted PET/CT scans, even using the novel SSTR-ligand [18F]SiTATE. Some inter-reader variability was observed regarding the evaluation of uptake intensity prior to PRRT as well as compartment scoring of lymph nodes, indicating that those categories require special attention during further clinical validation and might be refined in a future SSTR-RADS version 1.1. CLINICAL RELEVANCE STATEMENT SSTR-RADS 1.0 is a consistent framework for categorizing somatostatin receptor-targeted PET/CT scans when using [18F]SiTATE. The framework serves as a valuable tool for facilitating and improving the management of patients with NET. KEY POINTS SSTR-RADS 1.0 is a valuable tool for managing patients with NET. SSTR-RADS 1.0 categorizes patients with showing strong agreement across diverse reader expertise. As an alternative to [68Ga]-labeled PET/CT in neuroendocrine tumor imaging, SSTR-RADS 1.0 reliably classifies [18F]SiTATE-PET/CT.
Collapse
Affiliation(s)
- R Ebner
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany.
| | - A Lohse
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - M P Fabritius
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - J Rübenthaler
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), LMU University Hospital, LMU Munich, Munich, Germany
| | - C Wängler
- Biomedical Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany
| | - B Wängler
- Biomedical Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany
- Molecular Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany
| | - R Schirrmacher
- Department of Oncology, Division of Oncological Imaging, University of Alberta, Edmonton, Canada
| | - F Völter
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - H P Schmid
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - L M Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - O Öcal
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - A Hinterberger
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Heidelberg, Germany
| | - C Spitzweg
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), LMU University Hospital, LMU Munich, Munich, Germany
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - C J Auernhammer
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), LMU University Hospital, LMU Munich, Munich, Germany
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - T Geyer
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - J Ricke
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), LMU University Hospital, LMU Munich, Munich, Germany
| | - P Bartenstein
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), LMU University Hospital, LMU Munich, Munich, Germany
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - A Holzgreve
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - F Grawe
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Heidelberg, Germany
- Department of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
2
|
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
|
3
|
Zhang X, Choi JY, Lee KH, Choe YS. Synthesis and Evaluation of [ 18F]SiFA-Conjugated Ligands for Fibroblast Activation Protein Imaging. Mol Pharm 2023; 20:6441-6450. [PMID: 37968928 DOI: 10.1021/acs.molpharmaceut.3c00824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
In recent years, fibroblast activation protein (FAP) has emerged as an important target for the diagnosis and therapy of various tumors due to its high expression on the cell surface of cancer-associated fibroblasts, which are the major components of the tumor stroma. In this study, we synthesized and evaluated 18F-labeled FAP inhibitors (FAPIs) for FAP imaging. Two silicon fluoride acceptor (SiFA)-conjugated FAPIs were synthesized: one containing a γ-carboxy-l-glutamic acid (Gla) residue (1) and another containing two Gla residues (2). Both ligands exhibited high binding affinities for FAP. 18F/19F exchange reactions on both ligands were performed in the presence of 2% water. This resulted in the formation of radioligands [18F]1 and [18F]2 in high radiochemical yields. Radioligand [18F]2, with a more favorable partition coefficient, was selected for the U87MG cell binding study, and the results showed FAP-specific binding of the radioligand to the cells. An ex vivo biodistribution study in U87MG tumor-bearing mice 60 min after injection demonstrated a 5.8-fold higher tumor accumulation of [18F]2 than that of [18F]1. Furthermore, PET and ex vivo biodistribution studies of [18F]2 in U87MG tumor-bearing mice showed high and persistent tumor uptake over time, which was significantly blocked by the preinjection of FAPI-04. Our results indicate that [18F]SiFA-(Gla)2-conjugated FAPI ([18F]2) has the potential for FAP imaging.
Collapse
Affiliation(s)
- Xuran Zhang
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Joon Young Choi
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Kyung-Han Lee
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
| | - Yearn Seong Choe
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
| |
Collapse
|
4
|
Yang T, Peng L, Qiu J, He X, Zhang D, Wu R, Liu J, Zhang X, Zha Z. A radiohybrid theranostics ligand labeled with fluorine-18 and lutetium-177 for fibroblast activation protein-targeted imaging and radionuclide therapy. Eur J Nucl Med Mol Imaging 2023; 50:2331-2341. [PMID: 36864362 PMCID: PMC10250256 DOI: 10.1007/s00259-023-06169-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/19/2023] [Indexed: 03/04/2023]
Abstract
PURPOSE A series of radiotracers targeting fibroblast activation protein (FAP) with great pharmacokinetics have been developed for cancer diagnosis and therapy. Nevertheless, the use of dominant PET tracers, gallium-68-labeled FAPI derivatives, was limited by the short nuclide half-life and production scale, and the therapeutic tracers exhibited rapid clearance and insufficient tumor retention. In this study, we developed a FAP targeting ligand, LuFL, containing organosilicon-based fluoride acceptor (SiFA) and DOTAGA chelator, capable of labeling fluorine-18 and lutetium-177 in one molecular with simple and highly efficient labeling procedure, to achieve cancer theranostics. METHODS The precursor LuFL (20) and [natLu]Lu-LuFL (21) were successfully synthesized and labeled with fluorine-18 and lutetium-177 using a simple procedure. A series of cellular assays were performed to characterize the binding affinity and FAP specificity. PET imaging, SPECT imaging, and biodistribution studies were conducted to evaluate pharmacokinetics in HT-1080-FAP tumor-bearing nude mice. A comparison study of [177Lu]Lu-LuFL ([177Lu]21) and [177Lu]Lu-FAPI-04 was carried out in HT-1080-FAP xenografts to determine the cancer therapeutic efficacy. RESULTS LuFL (20) and [natLu]Lu-LuFL (21) demonstrated excellent binding affinity towards FAP (IC50: 2.29 ± 1.12 nM and 2.53 ± 1.87 nM), compared to that of FAPI-04 (IC50: 6.69 ± 0.88 nM). In vitro cellular studies showed that 18F-/177Lu-labeled 21 displayed high specific uptake and internalization in HT-1080-FAP cells. Micro-PET, SPECT imaging and biodistribution studies with [18F]/[177Lu]21 revealed higher tumor uptake and longer tumor retention than those of [68 Ga]/[177Lu]Ga/Lu-FAPI-04. The radionuclide therapy studies showed significantly greater inhibition of tumor growth for the [177Lu]21 group, than for the control group and the [177Lu]Lu-FAPI-04 group. CONCLUSION The novel FAPI-based radiotracer containing SiFA and DOTAGA was developed as a theranostics radiopharmaceutical with simple and short labeling process, and showed promising properties including higher cellular uptake, better FAP binding affinity, higher tumor uptake and prolong retention compared to FAPI-04. Preliminary experiments with 18F- and 177Lu-labeled 21 showed promising tumor imaging properties and favorable anti-tumor efficacy.
Collapse
Affiliation(s)
- Tianhong Yang
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, #58 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, China
| | - Lei Peng
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, #58 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, China
| | - Jia Qiu
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, #58 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, China
| | - Xingjin He
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, #58 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, China
| | - Dake Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, #58 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, China
| | - Renbo Wu
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, #58 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, China
| | - Jianbo Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, #58 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, China
| | - Xiangsong Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, #58 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, China.
| | - Zhihao Zha
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, #58 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, China.
| |
Collapse
|
5
|
Raheem SJ, Salih AK, Garcia MD, Sharpe JC, Toosi BM, Price EW. A Systematic Investigation into the Influence of Net Charge on the Biological Distribution of Radiometalated Peptides Using [ 68Ga]Ga-DOTA-TATE Derivatives. Bioconjug Chem 2023; 34:549-561. [PMID: 36800496 DOI: 10.1021/acs.bioconjchem.3c00007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Recently, several radiometalated peptides have been approved for clinical imaging and/or therapy (theranostics) of several types of cancer; nonetheless, the primary challenge that most of these peptides confront is significant renal uptake and retention, which is often dose limiting and can cause nephrotoxicity. In response to this, numerous methods have been employed to reduce the uptake of radiometalated peptides in the kidneys, and among these is adding a linker to modulate polarity and/or charge. To better understand the influence of net charge on the biodistribution of radiometalated peptides, we selected the clinically popular construct DOTA-TATE (NETSPOT/LUTATHERA) as a model system. We synthesized derivatives using manual solid-phase peptide synthesis methods including mechanical and ultrasonic agitation to effectively yield the gold standard DOTA-TATE and a series of derivatives with different net charges (+2, +1, 0, -1, -2). Dynamic PET imaging from 0 to 90 min in healthy female mice (CD1) revealed high accumulation and retention of activity in the kidneys for the net-neutral (0) charged [68Ga]Ga-DOTA-TATE and even higher for positively charged derivatives, whereas negatively charged derivatives exhibited low accumulation and fast renal excretion. Ex vivo biodistribution at 2 h post injection demonstrated a significant retention of [68Ga]Ga-DOTA-TATE (∼74 %ID/g) in the kidneys, which increased as the net positive charge per molecule increased to +1 and +2 (∼272 %ID/g and ∼333 %ID/g, respectively), but the -1 and -2 net charged molecules exhibited lower renal uptake (∼15 %ID/g and 16 %ID/g, respectively). Interestingly, the net -2 charged [68Ga]Ga-DOTA-(Glu)2-PEG4-TATE was stable in blood serum but had much higher healthy organ uptake (lungs, liver, spleen) than the net -1 compound, suggesting instability in vivo. Although the [68Ga]Ga-DOTA-PEG4-TATE derivative with a net charge of 0 also showed a decrease in kidney uptake, it also showed instability in blood serum and in vivo. Despite the superior pharmacokinetics of the net -1 charged [68Ga]Ga-DOTA-Glu-PEG4-TATE in healthy mice with respect to kidney uptake and overall profile, dynamic PET images and ex vivo biodistribution in male mice (NSG) bearing AR42J (SSTR2 overexpressing) subcutaneous tumor xenografts showed significantly diminished tumor uptake when compared to the gold standard [68Ga]Ga-DOTA-TATE. Taken together, these findings indicate unambiguously that kidney uptake and retention are significantly influenced by the net charge of peptide-based radiotracers. In addition, it was illustrated that the negatively charged peptides had substantially decreased kidney uptake, but in this instantiation the tumor uptake was also impaired.
Collapse
Affiliation(s)
- Shvan J Raheem
- Department of Chemistry, College of Arts and Science, University of Saskatchewan, 110 Science Place, S7N-5C9, Saskatoon, Saskatchewan, Canada
| | - Akam K Salih
- Department of Chemistry, College of Arts and Science, University of Saskatchewan, 110 Science Place, S7N-5C9, Saskatoon, Saskatchewan, Canada
| | - Moralba Dominguez Garcia
- Department of Chemistry, College of Arts and Science, University of Saskatchewan, 110 Science Place, S7N-5C9, Saskatoon, Saskatchewan, Canada
| | - Jessica C Sharpe
- Department of Small Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, S7N-5B4, Saskatoon, Saskatchewan, Canada
| | - Behzad M Toosi
- Department of Small Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, S7N-5B4, Saskatoon, Saskatchewan, Canada
| | - Eric W Price
- Department of Chemistry, College of Arts and Science, University of Saskatchewan, 110 Science Place, S7N-5C9, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
6
|
Efforts toward PET-Activatable Red-Shifted Silicon Rhodamines and Silicon Pyronine Dyes. Pharmaceuticals (Basel) 2023; 16:ph16030401. [PMID: 36986500 PMCID: PMC10053042 DOI: 10.3390/ph16030401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/14/2023] [Accepted: 02/18/2023] [Indexed: 03/09/2023] Open
Abstract
Tracers for bimodal optical imaging and positron emission tomography unite multiple advantages in a single molecule. Their tumor-specific uptake can be visualized after their PET activation by radiofluorination via PET/CT or PET/MRI allowing for staging or therapy planning, while their non-radioactive moiety additionally facilitates the visualization of malignant tissue during intraoperative fluorescence-guided surgery or in histological assessments. The silicon-bridged xanthene core offers the opportunity for radiofluorination with SiFA isotope exchange to obtain a small-molecule, PET-activatable NIR dye that can be linked to different target vectors. Herein, we demonstrate for the first time the PET-activation of a fluorinated silicon pyronine, belonging to a class of low-molecular-weight fluorescence dyes with a large Stokes shift (up to 129 nm) and solvent-dependent NIR dye properties, with a successful radiochemical conversion of 70%. The non-fluorinated pyronine precursor is easily accessible by a three-step sequence from commercially starting material with a 12% overall yield. Moreover, a library of seven unusually functionalized (by approximately 15 nm), red-shifted silicon rhodamines were synthesized in three- to four-step sequences and the optical properties of the novel dyes were characterized. It was also shown that the synthesized silicon rhodamine dyes can be easily conjugated by amide bond formation or ‘click-reaction’ approaches.
Collapse
|
7
|
Lozada J, Kuo HT, Lin WX, Lin KS, Bénard F, Perrin DM. Imidazolium-methylene-trifluoroborate: A novel radioprosthetic group validated with preclinical 18 F-Positron Emission Tomography imaging of Prostate Specific Membrane Antigen in mice. J Labelled Comp Radiopharm 2023; 66:130-137. [PMID: 36813569 DOI: 10.1002/jlcr.4020] [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: 12/03/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/24/2023]
Abstract
Organotrifluoroborates have gained acceptance as radioprosthetic groups for radiofluorination. Of these, the zwitterionic prosthetic group "AMBF3 " with a quaternary dimethylammonium ion dominates the trifluoroborate space. Herein, we report on imidazolium-methylene trifluoroborate (ImMBF3 ) as an alternative radioprosthetic group and report on its properties in the context of a PSMA-targeting EUK ligand that was previously been conjugated to AMBF3 . The ImMBF3 is readily synthesized from imidazole and conjugated via CuAAC "click" chemistry to give a structure similar to PSMA-617. 18 F-labeling proceeded in one step per our previous reports and imaged in LNCaP-xenograft bearing mice. The [18 F]-PSMA-617-ImMBF3 tracer proved to be less polar (LogP7.4 = -2.95 ± 0.03) while showing a significantly lower solvolytic rate (t1/2 = 8100 min) and slightly higher molar activity (Am) at 174 ± 38 GBq/μmol. Tumor uptake was measured at 13.7 ± 4.8%ID/g and a tumor:muscle ratio of 74.2 ± 35.0, tumor:blood ratio of 21.4 ± 7.0, tumor:kidney ratio of 0.29 ± 0.14, and tumor:bone ratio of 23.5 ± 9.5. In comparison with previously reported PSMA-targeting EUK-AMBF3 conjugates, we have altered the LogP7.4 value, tuned the solvolytic half-life of the prosthetic, and increased radiochemical conversion while achieving similar tumor uptake, contrast ratios, and molar activities compared with AMBF3 bioconjugates.
Collapse
Affiliation(s)
- Jerome Lozada
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hsiou-Ting Kuo
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Wen Xuan Lin
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kuo-Shyan Lin
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - François Bénard
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - David M Perrin
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
8
|
Abstract
18F-Labeling methods for the preparation of 18F-labeled molecular probes can be classified into electrophilic fluorination, nucleophilic fluorination, metal-F coordination, and 18F/19F isotope exchange. Isotope exchange-based 18F-labeling methods demonstrate mild conditions featuring water resistance and facile high-performance liquid chromatography-free purification in direct 18F-labeling of substrates. This paper systematically reviews isotope exchange-based 18F-labeling methods sorted by the adjacent atom bonding with F, i.e., carbon and noncarbon atoms (Si, B, P, S, Ga, Fe, etc.). The respective isotope exchange mechanism, radiolabeling condition, radiochemical yield, molar activity, and stability of the 18F-product are mainly discussed for each isotope exchange-based 18F-labeling method as well as the cutting-edge application of the corresponding 18F-labeled molecular probes.
Collapse
Affiliation(s)
- Tao Wang
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Experimental Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| | - Shengji Lv
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Experimental Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhaobiao Mou
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Experimental Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhenru Zhang
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Experimental Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| | - Taotao Dong
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Experimental Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| | - Zijing Li
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Experimental Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| |
Collapse
|
9
|
Bailey JJ, Wuest M, Wagner M, Bhardwaj A, Wängler C, Wängler B, Valliant JF, Schirrmacher R, Wuest F. Synthesis and Preclinical Evaluation of [ 18F]SiFA-PSMA Inhibitors in a Prostate Cancer Model. J Med Chem 2021; 64:15671-15689. [PMID: 34672630 DOI: 10.1021/acs.jmedchem.1c00812] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Positron emission tomography (PET) imaging of prostate-specific membrane antigen (PSMA) with gallium-68 (68Ga) and fluorine-18 (18F) radiotracers has aroused tremendous interest over the past few years. The use of organosilicon-[18F]fluoride acceptors (SiFA) conjugated to urea-based peptidomimetic PSMA inhibitors provides a "kit-like" multidose synthesis technology. Nine novel 18F-labeled SiFA-bearing PSMA inhibitors with different linker moieties were synthesized and analyzed for their in vitro binding against [125I]I-TAAG-PSMA in LNCaP cells. IC50 values ranged from 58-570 nM. Among all compounds, [18F]SiFA-Asp2-PEG3-PSMA (IC50 = 125 nM) showed the highest tumor uptake in LNCaP tumors (SUV60min 0.73). A substantial increase in molar activity (Am) (from 7.5 ± 0.5 to 86 ± 3 GBq/μmol) led to a significant increase in LNCaP tumor uptake (SUV60min 1.18; Δ 0.45 corresponding to +62%). In vivo blocking with DCFPyL resulted in -32% uptake after 60 min. The SiFA-isotopic exchange chemistry offers a method that is readily adaptable for a "kit-type" labeling procedure and clinical translation.
Collapse
Affiliation(s)
- Justin J Bailey
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Avenue, Edmonton, Alberta T6G 1Z2, Canada
| | - Melinda Wuest
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Avenue, Edmonton, Alberta T6G 1Z2, Canada
| | - Michael Wagner
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Avenue, Edmonton, Alberta T6G 1Z2, Canada
| | - Atul Bhardwaj
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Avenue, Edmonton, Alberta T6G 1Z2, Canada
| | - Carmen Wängler
- Clinic of Radiology and Nuclear Medicine, Biomedical Chemistry and Clinic of Radiology and Nuclear Medicine, Molecular Imaging and Radiochemistry, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim 68167, Germany
| | - Bjoern Wängler
- Clinic of Radiology and Nuclear Medicine, Biomedical Chemistry and Clinic of Radiology and Nuclear Medicine, Molecular Imaging and Radiochemistry, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim 68167, Germany
| | - John F Valliant
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main Street, Hamilton, Ontario L8S 4K1, Canada
| | - Ralf Schirrmacher
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Avenue, Edmonton, Alberta T6G 1Z2, Canada
| | - Frank Wuest
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Avenue, Edmonton, Alberta T6G 1Z2, Canada
| |
Collapse
|
10
|
Gower-Fry L, Kronemann T, Dorian A, Pu Y, Jaworski C, Wängler C, Bartenstein P, Beyer L, Lindner S, Jurkschat K, Wängler B, Bailey JJ, Schirrmacher R. Recent Advances in the Clinical Translation of Silicon Fluoride Acceptor (SiFA) 18F-Radiopharmaceuticals. Pharmaceuticals (Basel) 2021; 14:ph14070701. [PMID: 34358127 PMCID: PMC8309031 DOI: 10.3390/ph14070701] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/15/2021] [Accepted: 07/17/2021] [Indexed: 12/20/2022] Open
Abstract
The incorporation of silicon fluoride acceptor (SiFA) moieties into a variety of molecules, such as peptides, proteins and biologically relevant small molecules, has improved the generation of 18F-radiopharmaceuticals for medical imaging. The efficient isotopic exchange radiofluorination process, in combination with the enhanced [18F]SiFA in vivo stability, make it a suitable strategy for fluorine-18 incorporation. This review will highlight the clinical applicability of [18F]SiFA-labeled compounds and discuss the significant radiotracers currently in clinical use.
Collapse
Affiliation(s)
- Lexi Gower-Fry
- Department of Oncology, Division of Oncological Imaging, University of Alberta, Edmonton, AB T6G 1Z2, Canada; (L.G.-F.); (T.K.); (A.D.); (Y.P.); (C.J.); (J.J.B.)
| | - Travis Kronemann
- Department of Oncology, Division of Oncological Imaging, University of Alberta, Edmonton, AB T6G 1Z2, Canada; (L.G.-F.); (T.K.); (A.D.); (Y.P.); (C.J.); (J.J.B.)
| | - Andreas Dorian
- Department of Oncology, Division of Oncological Imaging, University of Alberta, Edmonton, AB T6G 1Z2, Canada; (L.G.-F.); (T.K.); (A.D.); (Y.P.); (C.J.); (J.J.B.)
| | - Yinglan Pu
- Department of Oncology, Division of Oncological Imaging, University of Alberta, Edmonton, AB T6G 1Z2, Canada; (L.G.-F.); (T.K.); (A.D.); (Y.P.); (C.J.); (J.J.B.)
| | - Carolin Jaworski
- Department of Oncology, Division of Oncological Imaging, University of Alberta, Edmonton, AB T6G 1Z2, Canada; (L.G.-F.); (T.K.); (A.D.); (Y.P.); (C.J.); (J.J.B.)
| | - Carmen Wängler
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany;
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany; (P.B.); (L.B.); (S.L.)
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany; (P.B.); (L.B.); (S.L.)
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany; (P.B.); (L.B.); (S.L.)
| | - Klaus Jurkschat
- Fakultät für Chemie und Chemische Biologie, Technische Universität Dortmund, 44227 Dortmund, Germany;
| | - Björn Wängler
- Molecular Imaging and Radiochemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany;
| | - Justin J. Bailey
- Department of Oncology, Division of Oncological Imaging, University of Alberta, Edmonton, AB T6G 1Z2, Canada; (L.G.-F.); (T.K.); (A.D.); (Y.P.); (C.J.); (J.J.B.)
| | - Ralf Schirrmacher
- Department of Oncology, Division of Oncological Imaging, University of Alberta, Edmonton, AB T6G 1Z2, Canada; (L.G.-F.); (T.K.); (A.D.); (Y.P.); (C.J.); (J.J.B.)
- Correspondence:
| |
Collapse
|
11
|
Automated synthesis of [ 18F]Ga-rhPSMA-7/ -7.3: results, quality control and experience from more than 200 routine productions. EJNMMI Radiopharm Chem 2021; 6:4. [PMID: 33484364 PMCID: PMC7826325 DOI: 10.1186/s41181-021-00120-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/11/2021] [Indexed: 12/15/2022] Open
Abstract
Introduction The radiohybrid (rh) prostate-specific membrane antigen (PSMA)-targeted ligand [18F]Ga-rhPSMA-7 has previously been clinically assessed and demonstrated promising results for PET-imaging of prostate cancer. The ligand is present as a mixture of four stereoisomers ([18F]Ga-rhPSMA-7.1, − 7.2, − 7.3 and − 7.4) and after a preclinical isomer selection process, [18F]Ga-rhPSMA-7.3 has entered formal clinical trials. Here we report on the establishment of a fully automated production process for large-scale production of [18F]Ga-rhPSMA-7/ -7.3 under GMP conditions (EudraLex). Methods [18F]Fluoride in highly enriched [18O]H2O was retained on a strong anion exchange cartridge, rinsed with anhydrous acetonitrile and subsequently eluted with a solution of [K+ ⊂ 2.2.2]OH− in anhydrous acetonitrile into a reactor containing Ga-rhPSMA ligand and oxalic acid in DMSO. 18F-for-19F isotopic exchange at the Silicon-Fluoride Acceptor (SiFA) was performed at room temperature, followed by dilution with buffer and cartridge-based purification. Optimum process parameters were determined on the laboratory scale and thereafter implemented into an automated synthesis. Data for radiochemical yield (RCY), purity and quality control were analyzed for 243 clinical productions (160 for [18F]Ga-rhPSMA-7; 83 for [18F]Ga-rhPSMA-7.3). Results The automated production of [18F]Ga-rhPSMA-7 and the single isomer [18F]Ga-rhPSMA-7.3 is completed in approx. 16 min with an average RCY of 49.2 ± 8.6% and an excellent reliability of 98.8%. Based on the different starting activities (range: 31–130 GBq, 89 ± 14 GBq) an average molar activity of 291 ± 62 GBq/μmol (range: 50–450 GBq/μmol) was reached for labeling of 150 nmol (231 μg) precursor. Radiochemical purity, as measured by radio-high performance liquid chromatography and radio-thin layer chromatography, was 99.9 ± 0.2% and 97.8 ± 1.0%, respectively. Conclusion This investigation demonstrates that 18F-for-19F isotopic exchange is well suited for the fast, efficient and reliable automated routine production of 18F-labeled PSMA-targeted ligands. Due to its simplicity, speed and robustness the development of further SiFA-based radiopharmaceuticals is highly promising and can be of far-reaching importance for future theranostic concepts. Supplementary Information The online version contains supplementary material available at 10.1186/s41181-021-00120-5.
Collapse
|
12
|
Mukai H, Watanabe Y. Review: PET imaging with macro- and middle-sized molecular probes. Nucl Med Biol 2021; 92:156-170. [PMID: 32660789 DOI: 10.1016/j.nucmedbio.2020.06.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 12/16/2022]
Abstract
Recent progress in radiolabeling of macro- and middle-sized molecular probes has been extending possibilities to use PET molecular imaging for dynamic application to drug development and therapeutic evaluation. Theranostics concept also accelerated the use of macro- and middle-sized molecular probes for sharpening the contrast of proper target recognition even the cellular types/subtypes and proper selection of the patients who should be treated by the same molecules recognition. Here, brief summary of the present status of immuno-PET, and then further development of advanced technologies related to immuno-PET, peptidic PET probes, and nucleic acids PET probes are described.
Collapse
Affiliation(s)
- Hidefumi Mukai
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| |
Collapse
|
13
|
Methods to radiolabel somatostatin analogs with [18F]fluoride: current status, challenges, and progress in clinical applications. J Radioanal Nucl Chem 2020. [DOI: 10.1007/s10967-020-07437-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
14
|
Raheem SJ, Schmidt BW, Solomon VR, Salih AK, Price EW. Ultrasonic-Assisted Solid-Phase Peptide Synthesis of DOTA-TATE and DOTA- linker-TATE Derivatives as a Simple and Low-Cost Method for the Facile Synthesis of Chelator-Peptide Conjugates. Bioconjug Chem 2020; 32:1204-1213. [PMID: 32645261 DOI: 10.1021/acs.bioconjchem.0c00325] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Peptides have been widely adopted as biological targeting vectors for applications in molecular imaging and peptide-receptor radionuclide therapy (PRRT). Somatostatin (SST) analogues such as octreotate (TATE) are exogenous ligands for somatostatin receptors (SSTRs), which are highly expressed on neuroendocrine tumors (NETs). Recently, both [68Ga]Ga-DOTA-TATE (NETSPOT) and [177Lu]Lu-DOTA-TATE (LUTATHERA) received U.S. Food and Drug Administration approval for positron emission tomography (PET) imaging and PRRT of NETs, respectively. However, to the best of our knowledge a well-described synthesis of DOTA-TATE has not been reported in the literature. Herein, we report a fully reoptimized DOTA-TATE synthesis, including the application of a simple ultrasonic bath to greatly improve yields, reduce coupling times, and decrease the amount of reagents required for each coupling step by a half. The most prevalently used cyclizing agents such as iodine, thallium(III) trifluoroacetate, hydrogen peroxide, and dimethyl sulfoxide were compared. On-resin cyclizations using mechanical agitation showed higher yields (23% and 25% using I2 and Tl(III), respectively) than off-resin (1.3% and 11% using DMSO and H2O2, respectively), and the total synthesis time of DOTA-TATE was ∼540 min excluding the cyclization step, with a total synthesis yield of ∼23%. The same manual SPPS methods/reagents were reoptimized with ultrasonic (US) agitation, resulting in an immense reduction in the total synthesis time by ∼8-fold to ∼70 min for DOTA-TATE with a higher yield (∼29% yield), and ∼13-fold to 105 min for DOTA-PEG4-TATE (∼29% yield). Also, the use of US agitation reduces the need for excess molar equivalents of the reagents to a half, which is particularly important when coupling expensive or custom-synthesized groups such as bifunctional chelators and linkers. Finally, the synthesized DOTA-TATE was successfully radiolabeled with [68Ga]Ga3+ (t1/2 = 68 min) with high radiochemical yields (30 min, 95 °C). We believe this work opens the door to the facile and low-cost synthesis of many new chelator-linker-peptide conjugates that were previously cumbersome or cost-prohibitive to produce with manual SPPS.
Collapse
Affiliation(s)
- Shvan J Raheem
- Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N-5C9, Canada
| | - Benjamin W Schmidt
- Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N-5C9, Canada
| | - Viswas Raja Solomon
- Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N-5C9, Canada
| | - Akam K Salih
- Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N-5C9, Canada
| | - Eric W Price
- Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N-5C9, Canada
| |
Collapse
|
15
|
Wurzer A, Di Carlo D, Schmidt A, Beck R, Eiber M, Schwaiger M, Wester HJ. Radiohybrid Ligands: A Novel Tracer Concept Exemplified by 18F- or 68Ga-Labeled rhPSMA Inhibitors. J Nucl Med 2020; 61:735-742. [PMID: 31862804 PMCID: PMC7198388 DOI: 10.2967/jnumed.119.234922] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 09/27/2019] [Indexed: 11/16/2022] Open
Abstract
When we critically assess the reason for the current dominance of 68Ga-labeled peptides and peptide-like ligands in radiopharmacy and nuclear medicine, we have to conclude that the major advantage of such radiopharmaceuticals is the apparent lack of suitable 18F-labeling technologies with proven clinical relevance. To prepare and to subsequently perform a clinical proof-of-concept study on the general suitability of silicon-fluoride-acceptor (SiFA)-conjugated radiopharmaceuticals, we developed inhibitors of the prostate-specific membrane antigen (PSMA) that are labeled by isotopic exchange (IE). To compensate for the pronounced lipophilicity of the SiFA unit, we used metal chelates, conjugated in close proximity to SiFA. Six different radiohybrid PSMA ligands (rhPSMA ligands) were evaluated and compared with the commonly used 18F-PSMA inhibitors 18F-DCFPyL and 18F-PSMA-1007. Methods: All inhibitors were synthesized by solid-phase peptide synthesis. Human serum albumin binding was measured by affinity high-performance liquid chromatography, whereas the lipophilicity of each tracer was determined by the n-octanol/buffer method. In vitro studies (IC50, internalization) were performed on LNCaP cells. Biodistribution studies were conducted on LNCaP tumor-bearing male CB-17 SCID mice. Results: On the laboratory scale (starting activities, 0.2-9.0 GBq), labeling of 18F-rhPSMA-5 to -10 by IE was completed in < 20 min (radiochemical yields, 58% ± 9%; radiochemical purity, >97%) with molar activities of 12-60 GBq/μmol. All rhPSMAs showed low nanomolar affinity and high internalization by PSMA-expressing cells when compared with the reference radiopharmaceuticals, medium-to-low lipophilicity, and high human serum albumin binding. Biodistribution studies in LNCaP tumor-bearing mice revealed high tumor uptake, sufficiently fast clearance kinetics from blood, low hepatobiliary excretion, fast renal excretion, and very low uptake of 18F activity in bone. Conclusion: The novel 18F-rhPSMA radiopharmaceuticals developed under the radiohybrid concept show equal or better targeting characteristics than the established 18F-PSMA tracers 18F-DCFPyL and 18F-PSMA-1007. The unparalleled simplicity of production, the possibility to produce the identical 68Ga-labeled 19F-68Ga-rhPSMA tracers, and the possibility to extend this concept to true theranostic radiohybrid radiopharmaceuticals, such as F-Lu-rhPSMA, are unique features of these radiopharmaceuticals.
Collapse
Affiliation(s)
- Alexander Wurzer
- Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany; and
| | - Daniel Di Carlo
- Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany; and
| | - Alexander Schmidt
- Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany; and
| | - Roswitha Beck
- Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany; and
| | - Matthias Eiber
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Hans-Jürgen Wester
- Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany; and
| |
Collapse
|
16
|
Abstract
Even so, the metal nanoparticles (metal NPs) have attractive optical and biomedical applications, the translation of metal NPs into the clinical practice remains a challenge due to their severe accumulation in the body. Active targeting to renal podocytes opens the door for enhancing kidney targeting and clearance. The goal of this study was to assess the excretion of larger particle size through kidney podocyte via active targeting. To reach this goal, PEGylated quantum dots (QDs) were coated with vapreotide (VAP) for selectively reaching somatostatin receptors (SSTRs) expressed in the podocyte cells. This QDs-VAP was tested on isolated primary podocytes, while the flow cytometry (FACS), confocal microscopy (CLSM), and inductively coupled plasma mass spectrometry (ICP-MS) were used to confirm this hypothesis. The results showed highly specific interactions with podocyte cells as detected by FACS, and CLSM. Moreover, ICP-MS demonstrated higher amount of QDs in the podocyte cells one-hour post-incubation (67.99% ID/g tissue), while the unmodified QDs did not accumulate. This study confirmed that QDs-VAP can target the podocyte's SSTRs then can be cleared via podocyte cells. Moreover, these results are considered as a highly promising approach for future therapy, targeting, clearance, and diagnosis of podocyte-associated diseases.
Collapse
Affiliation(s)
- Ahmed A H Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraydah 51452, Kingdom of Saudi Arabia.,Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| |
Collapse
|
17
|
Allott L, Dubash S, Aboagye EO. [ 18F]FET-βAG-TOCA: The Design, Evaluation and Clinical Translation of a Fluorinated Octreotide. Cancers (Basel) 2020; 12:cancers12040865. [PMID: 32252406 PMCID: PMC7226534 DOI: 10.3390/cancers12040865] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/26/2020] [Accepted: 03/31/2020] [Indexed: 01/17/2023] Open
Abstract
The success of Lutathera™ ([177Lu]Lu-DOTA-TATE) in the NETTER-1 clinical trial as a peptide receptor radionuclide therapy (PRRT) for somatostatin receptor expressing (SSTR) neuroendocrine tumours (NET) is likely to increase the demand for patient stratification by positron emission tomography (PET). The current gold standard of gallium-68 radiolabelled somatostatin analogues (e.g., [68Ga]Ga-DOTA-TATE) works effectively, but access is constrained by the limited availability and scalability of gallium-68 radiopharmaceutical production. The aim of this review is three-fold: firstly, we discuss the peptide library design, biological evaluation and clinical translation of [18F]fluoroethyltriazole-βAG-TOCA ([18F]FET-βAG-TOCA), our fluorine-18 radiolabelled octreotide; secondly, to exemplify the potential of the 2-[18F]fluoroethylazide prosthetic group and copper-catalysed azide-alkyne cycloaddition (CuAAC) chemistry in accessing good manufacturing practice (GMP) compatible radiopharmaceuticals; thirdly, we aim to illustrate a framework for the translation of similarly radiolabelled peptides, in which in vivo pharmacokinetics drives candidate selection, supported by robust radiochemistry methodology and a route to GMP production. It is hoped that this review will continue to inspire the development and translation of fluorine-18 radiolabelled peptides into clinical studies for the benefit of patients.
Collapse
|
18
|
Narayanam MK, Toutov AA, Murphy JM. Rapid One-Step 18F-Labeling of Peptides via Heteroaromatic Silicon-Fluoride Acceptors. Org Lett 2020; 22:804-808. [DOI: 10.1021/acs.orglett.9b04160] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Maruthi Kumar Narayanam
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
| | - Anton A. Toutov
- Fuzionaire Diagnostics, Inc., 177 East Colorado Boulevard, Pasadena, California 91105, United States
| | - Jennifer M. Murphy
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
| |
Collapse
|
19
|
Waldmann CM, Stuparu AD, van Dam RM, Slavik R. The Search for an Alternative to [ 68Ga]Ga-DOTA-TATE in Neuroendocrine Tumor Theranostics: Current State of 18F-labeled Somatostatin Analog Development. Theranostics 2019; 9:1336-1347. [PMID: 30867834 PMCID: PMC6401503 DOI: 10.7150/thno.31806] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/14/2019] [Indexed: 02/06/2023] Open
Abstract
The trend to inform personalized molecular radiotherapy with molecular imaging diagnostics, a concept referred to as theranostics, has transformed the field of nuclear medicine in recent years. The development of theranostic pairs comprising somatostatin receptor (SSTR)-targeting nuclear imaging probes and therapeutic agents for the treatment of patients with neuroendocrine tumors (NETs) has been a driving force behind this development. With the Neuroendocrine Tumor Therapy (NETTER-1) phase 3 trial reporting encouraging results in the treatment of well-differentiated, metastatic midgut NETs, peptide radioligand therapy (RLT) with the 177Lu-labeled somatostatin analog (SSA) [177Lu]Lu-DOTA-TATE is now anticipated to become the standard of care. On the diagnostics side, the field is currently dominated by 68Ga-labeled SSAs for the molecular imaging of NETs with positron emission tomography-computed tomography (PET/CT). PET/CT imaging with SSAs such as [68Ga]Ga-DOTA-TATE, [68Ga]Ga-DOTA-TOC, and [68Ga]Ga-DOTA-NOC allows for NET staging with high accuracy and is used to qualify patients for RLT. Driven by the demand for PET/CT imaging of NETs, a commercial kit for the production of [68Ga]Ga-DOTA-TATE (NETSPOT) was approved by the U.S. Food and Drug Administration (FDA). The synthesis of 68Ga-labeled SSAs from a 68Ge/68Ga-generator is straightforward and allows for a decentralized production, but there are economic and logistic difficulties associated with these approaches that warrant the search for a viable, generator-independent alternative. The clinical introduction of an 18F-labeled SSTR-imaging probe can help mitigate the shortcomings of the generator-based synthesis approach, but despite extensive research efforts, none of the proposed 18F-labeled SSAs has been translated past prospective first-in-humans studies so far. Here, we review the current state of probe-development from a translational viewpoint and make a case for a clinically viable, 18F-labeled alternative to the current standard [68Ga]Ga-DOTA-TATE.
Collapse
Affiliation(s)
- Christopher M. Waldmann
- Ahmanson Translational Imaging Division, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Andreea D. Stuparu
- Ahmanson Translational Imaging Division, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - R. Michael van Dam
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Roger Slavik
- Ahmanson Translational Imaging Division, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| |
Collapse
|
20
|
Illert P, Wängler B, Wängler C, Zöllner F, Uhrig T, Litau S, Pretze M, Röder T. Functionalizable composite nanoparticles as a dual magnetic resonance imaging/computed tomography contrast agent for medical imaging. J Appl Polym Sci 2019. [DOI: 10.1002/app.47571] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Patrick Illert
- Institute of Chemical Process EngineeringMannheim University of Applied Sciences Paul‐Wittsack‐Street 10, 68163 Mannheim Germany
| | - Björn Wängler
- Molecular Imaging and Radiochemistry, Department of Clinical Radiology and Nuclear MedicineMedical Faculty Mannheim of Heidelberg University Theodor‐Kutzer‐Ufer 1‐3, 68167 Mannheim Germany
| | - Carmen Wängler
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear MedicineMedical Faculty Mannheim of Heidelberg University Theodor‐Kutzer‐Ufer 1‐3, 68167 Mannheim Germany
| | - Frank Zöllner
- Computer Assisted Clinical Medicine, Medical Faculty Mannheim of Heidelberg University Theodor‐Kutzer‐Ufer 1‐3, 68167 Mannheim Germany
| | - Tanja Uhrig
- Computer Assisted Clinical Medicine, Medical Faculty Mannheim of Heidelberg University Theodor‐Kutzer‐Ufer 1‐3, 68167 Mannheim Germany
| | - Shanna Litau
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear MedicineMedical Faculty Mannheim of Heidelberg University Theodor‐Kutzer‐Ufer 1‐3, 68167 Mannheim Germany
| | - Marc Pretze
- Molecular Imaging and Radiochemistry, Department of Clinical Radiology and Nuclear MedicineMedical Faculty Mannheim of Heidelberg University Theodor‐Kutzer‐Ufer 1‐3, 68167 Mannheim Germany
| | - Thorsten Röder
- Institute of Chemical Process EngineeringMannheim University of Applied Sciences Paul‐Wittsack‐Street 10, 68163 Mannheim Germany
| |
Collapse
|
21
|
Abdellatif AA, Abou-Taleb HA, Abd El Ghany AA, Lutz I, Bouazzaoui A. Targeting of somatostatin receptors expressed in blood cells using quantum dots coated with vapreotide. Saudi Pharm J 2018; 26:1162-1169. [PMID: 30532637 PMCID: PMC6260484 DOI: 10.1016/j.jsps.2018.07.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 07/19/2018] [Indexed: 12/27/2022] Open
Abstract
Cancer may be difficult to target, however, if cancer targeted this provides the chance for a better and more effective treatment. Quantum dots (Qdots) coated vapreotide (VAP) as a somatostatin receptors (SSTRs) agonist can be efficient targeting issue since may reduce side effects and increase drug delivery to the target tissue. This study highlights the active targeting of cancer cells by cells imaging with improving the therapeutic outcomes. VAP was conjugated to Qdots using amine-to-sulfhydryl crosslinker. The synthesized Qdots-VAP was characterized by determination of size, measuring the zeta-potential and UV fluorometer. The cellular uptake was studied using different cell lines. Finally, the Qdots-VAP was injected into a rat model. The results showed a size of 479.8 ± 15 and 604.88 ± 17 nm for unmodified Qdots and Qdots-VAP respectively, while the zeta potential of particles went from negative to positive charge which proved the conjugation of VAP to Qdots. The fluorometer recorded a redshift for Qdots-VAP compared with unmodified Qdots. Moreover, cellular uptake exhibited high specific binding with cells which express SSTRs using confocal microscopy and flow cytometry (17.3 MFU comparing to 3.1 MFU of control, P < 0.001). Finally, an in vivo study showed a strong accumulation of Qdots-VAP in the blood cells (70%). In conclusion, Qdots-VAP can play a crucial role in cancer diagnosis and treatment of blood cells diseases when conjugated with VAP as SSTRs agonist.
Collapse
Affiliation(s)
- Ahmed A.H. Abdellatif
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Al-Azhar University, 71524 Assiut, Egypt
- Pharmaceutics Department, Faculty of Pharmacy, Qassim University, 51452 Buraydah, Saudi Arabia
| | - Heba A. Abou-Taleb
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Nahda University (NUB), Benisuef, Egypt
| | | | - Ilka Lutz
- Leibniz-Institute of Freshwater Ecology and Inland Fisheries, Mueggelseedamm 301, 12587 Berlin, Germany
| | - Abdellatif Bouazzaoui
- Science and Technology Unit, Umm Al Qura University, Makkah 21955, Saudi Arabia
- Internal Medicine 3-Hematology/Oncology Department, University Medical Center, Regensburg, Germany
| |
Collapse
|
22
|
Perrin DM. Organotrifluoroborates as prosthetic groups for Single-Step F18-Labeling of Complex Molecules. Curr Opin Chem Biol 2018; 45:86-94. [DOI: 10.1016/j.cbpa.2018.03.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 02/23/2018] [Accepted: 03/07/2018] [Indexed: 12/11/2022]
|
23
|
Abstract
Hydrolytic enzymes are a large class of biological catalysts that play a vital role in a plethora of critical biochemical processes required to maintain human health. However, the expression and/or activity of these important enzymes can change in many different diseases and therefore represent exciting targets for the development of positron emission tomography (PET) and single-photon emission computed tomography (SPECT) radiotracers. This review focuses on recently reported radiolabeled substrates, reversible inhibitors, and irreversible inhibitors investigated as PET and SPECT tracers for imaging hydrolytic enzymes. By learning from the most successful examples of tracer development for hydrolytic enzymes, it appears that an early focus on careful enzyme kinetics and cell-based studies are key factors for identifying potentially useful new molecular imaging agents.
Collapse
Affiliation(s)
- Brian P Rempel
- 1 Department of Science, Augustana Faculty, University of Alberta, Edmonton, Alberta, Canada
| | - Eric W Price
- 2 Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Christopher P Phenix
- 2 Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,3 Biomarker Discovery, Thunder Bay Regional Health Research Institute, Thunder Bay, Ontario, Canada
| |
Collapse
|
24
|
Vall-Sagarra A, Litau S, Decristoforo C, Wängler B, Schirrmacher R, Fricker G, Wängler C. Design, Synthesis, In Vitro, and Initial In Vivo Evaluation of Heterobivalent Peptidic Ligands Targeting Both NPY(Y₁)- and GRP-Receptors-An Improvement for Breast Cancer Imaging? Pharmaceuticals (Basel) 2018; 11:ph11030065. [PMID: 29973529 PMCID: PMC6161111 DOI: 10.3390/ph11030065] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 06/29/2018] [Accepted: 07/03/2018] [Indexed: 12/20/2022] Open
Abstract
Heterobivalent peptidic ligands (HBPLs), designed to address two different receptors independently, are highly promising tumor imaging agents. For example, breast cancer has been shown to concomitantly and complementarily overexpress the neuropeptide Y receptor subtype 1 (NPY(Y1)R) as well as the gastrin-releasing peptide receptor (GRPR). Thus, radiolabeled HBPLs being able to bind these two receptors should exhibit an improved tumor targeting efficiency compared to monospecific ligands. We developed here such bispecific HBPLs and radiolabeled them with 68Ga, achieving high radiochemical yields, purities, and molar activities. We evaluated the HBPLs and their monospecific reference peptides in vitro regarding stability and uptake into different breast cancer cell lines and found that the 68Ga-HBPLs were efficiently taken up via the GRPR. We also performed in vivo PET/CT imaging and ex vivo biodistribution studies in T-47D tumor-bearing mice for the most promising 68Ga-HBPL and compared the results to those obtained for its scrambled analogs. The tumors could easily be visualized by the newly developed 68Ga-HBPL and considerably higher tumor uptakes and tumor-to-background ratios were obtained compared to the scrambled analogs in and ex vivo. These results demonstrate the general feasibility of the approach to use bispecific radioligands for in vivo imaging of breast cancer.
Collapse
Affiliation(s)
- Alicia Vall-Sagarra
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany.
| | - Shanna Litau
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany.
- Molecular Imaging and Radiochemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany.
| | - Clemens Decristoforo
- Department of Nuclear Medicine, University Hospital Innsbruck, Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria.
| | - Björn Wängler
- Molecular Imaging and Radiochemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany.
| | - Ralf Schirrmacher
- Department of Oncology, Division Oncological Imaging, University of Alberta, 11560 University Avenue, Edmonton, AB T6G 1Z2, Canada.
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany.
| | - Carmen Wängler
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany.
| |
Collapse
|
25
|
Lindner S, Fiedler L, Wängler B, Bartenstein P, Schirrmacher R, Wängler C. Design, synthesis and in vitro evaluation of heterobivalent peptidic radioligands targeting both GRP- and VPAC1-Receptors concomitantly overexpressed on various malignancies – Is the concept feasible? Eur J Med Chem 2018; 155:84-95. [DOI: 10.1016/j.ejmech.2018.05.047] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 05/23/2018] [Accepted: 05/28/2018] [Indexed: 02/06/2023]
|
26
|
Kumar K, Ghosh A. 18F-AlF Labeled Peptide and Protein Conjugates as Positron Emission Tomography Imaging Pharmaceuticals. Bioconjug Chem 2018; 29:953-975. [PMID: 29463084 DOI: 10.1021/acs.bioconjchem.7b00817] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The clinical applications of positron emission tomography (PET) imaging pharmaceuticals have increased tremendously over the past several years since the approval of 18fluorine-fluorodeoxyglucose (18F-FDG) by the Food and Drug Administration (FDA). Numerous 18F-labeled target-specific potential imaging pharmaceuticals, based on small and large molecules, have been evaluated in preclinical and clinical settings. 18F-labeling of organic moieties involves the introduction of the radioisotope by C-18F bond formation via a nucleophilic or an electrophilic substitution reaction. However, biomolecules, such as peptides, proteins, and oligonucleotides, cannot be radiolabeled via a C-18F bond formation as these reactions involve harsh conditions, including organic solvents, high temperature, and nonphysiological conditions. Several approaches, including 18F-labeled prosthetic groups, silicon, boron, and aluminum fluoride acceptor chemistry, and click chemistry have been developed, in the past, for 18F labeling of biomolecules. Linear and macrocyclic polyaminocarboxylates and their analogs and derivatives form thermodynamically stable and kinetically inert aluminum chelates. Hence, macrocyclic polyaminocarboxylates have been used for conjugation with biomolecules, such as folate, peptides, affibodies, and protein fragments, followed by 18F-AlF chelation, and evaluation of their targeting abilities in preclinical and clinical environments. The goal of this report is to provide an overview of the 18F radiochemistry and 18F-labeling methodologies for small molecules and target-specific biomolecules, a comprehensive review of coordination chemistry of Al3+, 18F-AlF labeling of peptide and protein conjugates, and evaluation of 18F-labeled biomolecule conjugates as potential imaging pharmaceuticals.
Collapse
Affiliation(s)
- Krishan Kumar
- Laboratory for Translational Research in Imaging Pharmaceuticals, The Wright Center of Innovation in Biomedical Imaging, Department of Radiology , The Ohio State University , Columbus , Ohio 43212 , United States
| | - Arijit Ghosh
- Laboratory for Translational Research in Imaging Pharmaceuticals, The Wright Center of Innovation in Biomedical Imaging, Department of Radiology , The Ohio State University , Columbus , Ohio 43212 , United States
| |
Collapse
|
27
|
Perrio C, Schmitt S, Pla D, Gabbaï FP, Chansaenpak K, Mestre-Voegtle B, Gras E. [18F]-Fluoride capture and release: azeotropic drying free nucleophilic aromatic radiofluorination assisted by a phosphonium borane. Chem Commun (Camb) 2017; 53:340-343. [DOI: 10.1039/c6cc05168e] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
[18F]-Fluoride ready for SNAr was prepared according to a simple process including trapping of aqueous [18F]-fluoride on a cartridge pre-loaded with the phosphonium borane [(Ph2MeP)C6H4(BMes2)]+, then releasing by elution of TBACN in dry acetonitrile.
Collapse
Affiliation(s)
| | | | - Daniel Pla
- LCC
- CNRS
- UPR8241
- UFTMP
- 31077 Toulouse Cedex 4
| | | | | | | | | |
Collapse
|
28
|
Maschauer S, Heilmann M, Wängler C, Schirrmacher R, Prante O. Radiosynthesis and Preclinical Evaluation of 18F-Fluoroglycosylated Octreotate for Somatostatin Receptor Imaging. Bioconjug Chem 2016; 27:2707-2714. [PMID: 27715017 DOI: 10.1021/acs.bioconjchem.6b00472] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Short synthetic octapeptide analogs derived from the native somatostatin peptides SST-14 and SST-28, namely, octreotate (TATE) or octreotide (TOC), bind with high affinity to somatostatin receptors (sstr), mainly to subtypes 2 and 5, which are expressed in high density on neuroendocrine tumors (NET). Therefore, radiolabeled TATE or TOC derivatives represent highly valuable imaging probes for NET diagnosis by positron emission tomography (PET). The aim of our study was the development of an 18F-labeled octreotate analog as an alternative radiotracer for the clinically established 68Ga-DOTATOC and 68Ga-DOTATATE. We applied our previously developed method based on copper(I)-catalyzed azide-alkyne cycloaddition (CuAAC) to the radiosynthesis of 18F-fluoroglycosylated TATE ([18F]FGlc-TATE). [18F]FGlc-TATE was obtained in high yields of 19-22% (non-decay-corrected, referred to [18F]fluoride) and in high specific activities of 32-106 GBq/μmol. [18F]FGlc-TATE showed high affinity to sstr expressed on AR42J cells (IC50 = 4.2 nM) with fast and high internalization, and a beneficial logD7.4 of -1.8. In AR42J tumor bearing nude mice, [18F]FGlc-TATE showed high and specific tumor uptake of 5.6%ID/g at 60 min post-injection, as determined by blocking experiments using octreotide, and fast clearance from other organs, resulting in excellent tumor-to-blood ratios increasing from 9 to 17 from 30 to 60 min post-injection. Small animal PET studies revealed high uptake of [18F]FGlc-TATE in the tumor which could be blocked with octreotide by >99%. Overall, [18F]FGlc-TATE revealed excellent in vitro and in vivo properties and is therefore a viable alternative 18F-labeled radiopeptide for imaging somatostatin receptor-positive tumors by PET.
Collapse
Affiliation(s)
- Simone Maschauer
- Molecular Imaging and Radiochemistry, Department of Nuclear Medicine, Friedrich Alexander University Erlangen-Nürnberg (FAU) , Schwabachanlage 6, 91054 Erlangen, Germany
| | - Marcus Heilmann
- Molecular Imaging and Radiochemistry, Department of Nuclear Medicine, Friedrich Alexander University Erlangen-Nürnberg (FAU) , Schwabachanlage 6, 91054 Erlangen, Germany
| | - Carmen Wängler
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear Medicine Medical Faculty Mannheim of Heidelberg University , Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Ralf Schirrmacher
- Medical Isotope Cyclotron Facility, Department of Oncology, University of Alberta , 11560 University Avenue, Edmonton, Alberta T6G 1Z2, Canada
| | - Olaf Prante
- Molecular Imaging and Radiochemistry, Department of Nuclear Medicine, Friedrich Alexander University Erlangen-Nürnberg (FAU) , Schwabachanlage 6, 91054 Erlangen, Germany
| |
Collapse
|
29
|
Charron CL, Farnsworth AL, Roselt PD, Hicks RJ, Hutton CA. Recent developments in radiolabelled peptides for PET imaging of cancer. Tetrahedron Lett 2016. [DOI: 10.1016/j.tetlet.2016.07.083] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
30
|
Poschenrieder A, Osl T, Schottelius M, Hoffmann F, Wirtz M, Schwaiger M, Wester HJ. First 18F-Labeled Pentixafor-Based Imaging Agent for PET Imaging of CXCR4 Expression In Vivo. ACTA ACUST UNITED AC 2016; 2:85-93. [PMID: 30042959 PMCID: PMC6024415 DOI: 10.18383/j.tom.2016.00130] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In vivo quantification of CXCR4 expression using [68Ga]pentixafor for positron emission tomography (PET) imaging has gained significant clinical interest as CXCR4 plays a fundamental role in oncology and possesses potential prognostic value when overexpressed. To combine the excellent CXCR4-targeting properties of pentixafor-based tracers with the favorable radionuclide properties of 18F for high-resolution PET imaging, we developed an Al18F-labeled 1,4,7-triazacyclononane-triacetic acid (NOTA) analog of pentixather. Al18F-labeling of NOTA-pentixather was performed in aqueous dimethyl sulfoxide (DMSO) at pH = 4 (105°C, 15 minutes). CXCR4 affinities were determined in competitive binding assays, and both biodistribution and small-animal PET studies were performed in Daudi lymphoma-bearing mice. Under non-optimized conditions, [18F]AlF-NOTA-pentixather was obtained in radiochemical yields of 45.5% ± 13.3% and specific activities of up to 24.8 GBq/μmol. Compared with [natGa]pentixafor, [natF]AlF-NOTA-pentixather showed 1.4-fold higher CXCR4 affinity. [18F]AlF-NOTA-pentixather displayed high and CXCR4-specific in vivo uptake in Daudi xenografts (13.9% ± 0.8% injected dose per gram [ID/g] at 1 hour post injection [p.i.]). Because of its enhanced lipophilicity (logP = -1.4), [18F]AlF-NOTA-pentixather showed increased accumulation in the gall bladder and intestines. However, tumor/background ratios of 7.0 ± 1.2, 2.0 ± 0.3, 2.2 ± 0.4, 16.5 ± 6.5, and 29.2 ± 4 for blood, liver, small intestine, gut, and muscle, respectively, allowed for high-contrast visualization of Daudi tumors using PET (1 hour p.i.). The relatively straightforward radiosynthesis and efficient CXCR4 targeting of [18F]AlF-NOTA-pentixather demonstrate the successful implementation of 18F-complexation chemistry and pentixather-based CXCR4 targeting. Upon pharmacokinetic optimization, this class of tracers holds great promise for future application in humans.
Collapse
Affiliation(s)
| | - Theresa Osl
- Pharmaceutical Radiochemistry, Technische Universität München, Germany and
| | | | - Frauke Hoffmann
- Pharmaceutical Radiochemistry, Technische Universität München, Germany and
| | - Martina Wirtz
- Pharmaceutical Radiochemistry, Technische Universität München, Germany and
| | - Markus Schwaiger
- Nuklearmedizinische Klinik und Poliklinik, Technische Universität München, Ismaningerstr, München, Germany
| | - Hans-Jürgen Wester
- Pharmaceutical Radiochemistry, Technische Universität München, Germany and
| |
Collapse
|
31
|
Bernard-Gauthier V, Bailey JJ, Liu Z, Wängler B, Wängler C, Jurkschat K, Perrin DM, Schirrmacher R. From Unorthodox to Established: The Current Status of (18)F-Trifluoroborate- and (18)F-SiFA-Based Radiopharmaceuticals in PET Nuclear Imaging. Bioconjug Chem 2015; 27:267-79. [PMID: 26566577 DOI: 10.1021/acs.bioconjchem.5b00560] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Unorthodox (18)F-labeling strategies not employing the formation of a carbon-(18)F bond are seldom found in radiochemistry. Historically, the formation of a boron- or silicon-(18)F bond has been introduced very early on into the repertoire of labeling chemistries, but is without translation into any clinical radiotracer besides inorganic B[(18)F]F4(-) for brain tumor diagnosis. For many decades these labeling methodologies were forgotten and have just recently been revived by a handful of researchers thinking outside the box. When breaking with established paradigms such as the inability to obtain labeled compounds of high specific activity via isotopic exchange or performing radiofluorination in aqueous media, the research community often reacts skeptically. In 2005 and 2006, two novel labeling methodologies were introduced into radiochemistry for positron emission tomography (PET) tracer development: RBF3(-) labeling reported by Perrin et al. and the SiFA methodology by Schirrmacher, Jurkschat, and Waengler et al. which is based on isotopic exchange (IE). Both labeling methodologies have been complemented by other noncanonical strategies to introduce (18)F into biomolecules of diagnostic importance, thus profoundly enriching the landscape of (18)F radiolabeling. B- and Si-based labeling strategies finally revealed that IE is a viable alternative to established and traditional radiochemistry with the advantage of simplifying both the labeling effort as well as the necessary purification of the radiotracer. Hence IE will be the focus of this contribution over other noncanonical labeling methods. Peptides for tumor imaging especially lend themselves favorably toward one-step labeling via IE, but small molecules have been described as well, taking advantage of these new approaches, and have been used successfully for brain imaging. This Review gives an account of both radiochemistries centered on boron and silicon, describing the very beginnings of their basic research, the path that led to optimization of their chemistries, and the first encouraging preclinical results paving the way to their clinical use. This side by side approach will give the reader the opportunity to follow the development of a new basic discovery into a clinically applicable radiotracer including all the hurdles that have had to be overcome.
Collapse
Affiliation(s)
- Vadim Bernard-Gauthier
- Division of Oncological Imaging, Department of Oncology, University of Alberta , 11560 University Avenue, Edmonton, Alberta T6G 1Z2, Canada
| | - Justin J Bailey
- Division of Oncological Imaging, Department of Oncology, University of Alberta , 11560 University Avenue, Edmonton, Alberta T6G 1Z2, Canada
| | - Zhibo Liu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health , 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | | | | | - Klaus Jurkschat
- Department of Chemistry and Chemical Biology, Technical University of Dortmund , 44227 Dortmund, Germany
| | - David M Perrin
- Department of Chemistry, University of British Columbia , 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | - Ralf Schirrmacher
- Division of Oncological Imaging, Department of Oncology, University of Alberta , 11560 University Avenue, Edmonton, Alberta T6G 1Z2, Canada
| |
Collapse
|