1
|
Huang W, Wen K, Muschitiello PF, Escorihuela J, Laughlin ST. Evaluation of Pyrones in Bioorthogonal Reactions: Correlation between Structure, Reactivity, and Bioorthogonality. J Org Chem 2025; 90:2848-2859. [PMID: 39967512 DOI: 10.1021/acs.joc.4c02336] [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: 02/20/2025]
Abstract
Alpha-pyrones have been used for applications ranging from total synthesis to antibiotics. However, their application as dienes in bioorthogonal reactions has not been extensively explored. In previous work, we demonstrated the promising application of ester-functionalized pyrones in bioorthogonal protein labeling. Here, we constructed a library of substituted pyrones to evaluate their potential in bioorthogonal reactions by exploring the relationships among structure, reactivity, and bioorthogonality. We found that most pyrone derivatives with electron-withdrawing groups exhibited reactivity toward endo-bicyclo[6.1.0]nonyne (BCN), producing tricyclic and tetracyclic products in good yields. As expected, pyrones with more and stronger electron-withdrawing substituents showed faster reaction kinetics with BCN. Bicyclic pyrone derivatives showed substantially decreased reactivity, most likely resulting from increased steric effects. Counterintuitively, we found that substitutions at pyrone positions 4 and 5 affected the reactivity more than those at positions 3 and 6. To provide insights into both the expected and counterintuitive reactivities of the pyrone library members, we performed a quantum chemical analysis. Additionally, we evaluated each pyrone's reactivity with L-cysteine and found no correlation between pyrone reactivity with BCN and cysteine-based bioorthogonality. Finally, we evaluated the reactivity of pyrones toward a collection of popular dienophiles used in bioorthogonal reactions.
Collapse
Affiliation(s)
- Wei Huang
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11790, United States
| | - Kangqiao Wen
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11790, United States
| | - Paul F Muschitiello
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11790, United States
| | - Jorge Escorihuela
- Departamento de Química Orgánica, Universitat de València, Burjassot 46100, Valencia, Spain
| | - Scott T Laughlin
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11790, United States
| |
Collapse
|
2
|
Diao W, Lu J, Li J, Wu X, Cai H, He X, Pan L, Cheng Z, Wu H, Jia Z, Mao W. Cysteine-Specific 18F and NIR Dual Labeling of Peptides via Vinyltetrazine Bioorthogonal Conjugation for Molecular Imaging. J Med Chem 2025; 68:1526-1539. [PMID: 39754584 DOI: 10.1021/acs.jmedchem.4c02165] [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: 01/06/2025]
Abstract
Radiolabeled peptides are vital for positron emission tomography (PET) imaging, yet the 18F-labeling peptides remain challenging due to harsh conditions and time-consuming premodification requirements. Herein, we developed a novel vinyltetrazine-mediated bioorthogonal approach for highly efficient 18F-radiolabeling of a native peptide under mild conditions. This approach enabled radiosynthesis of various tumor-targeting PET tracers, including targeting the neurofibromin receptor (18F-P10a), the integrin αvβ3 (18F-P12a), and the platelet-derived growth factor receptor β (18F-ZPDGFRβ), with a radiochemical yield exceeding 90%. Preliminary evaluations revealed excellent hydrophilicity across these tracers, with 18F-P12a effectively visualizing integrin αvβ3 expression (tumor uptake: 1.57 ± 0.54%ID/g at 2 h). Additionally, we explored the potential for development of PET/near-infrared (NIR) dual-labeling agents using this method. The dual-modality agent 18F-Cy5-P12d enables specificity and colocalized imaging integrin αvβ3 expression (tumor uptake: 1.35 ± 0.24%ID/g at 2 h). Overall, this strategy offers a versatile platform for peptide radiolabeling and dual-modality agent development.
Collapse
Affiliation(s)
- Wei Diao
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
- Department of Nuclear Medicine, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Elctronic Science and Technology of China, Chengdu 610041, China
| | - Jing Lu
- Medical Insurance Office, West China Hospital of Sichuan University, Chengdu 610041, China
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Li
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Frontiers Science Center for Disease Related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoai Wu
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Huawei Cai
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xinyu He
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Frontiers Science Center for Disease Related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lili Pan
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Zhuzhong Cheng
- Department of Nuclear Medicine, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Elctronic Science and Technology of China, Chengdu 610041, China
| | - Haoxing Wu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Frontiers Science Center for Disease Related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiyun Jia
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| | - Wuyu Mao
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
3
|
Yang C, Lu K, Li J, Wu H, Chen W. Rapid Construction of 18F-Triazolyl-tetrazines through the Click Reaction. J Org Chem 2024; 89:14673-14678. [PMID: 38875503 DOI: 10.1021/acs.joc.4c00574] [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: 06/16/2024]
Abstract
Due to the fast reaction rate, 18F-labeled tetrazines have been widely applied in positron emission tomography (PET) imaging in cancer research and drug discovery. In this work, several functional 18F-triazolyl-tetrazines were rapidly obtained through an optimized copper-catalyzed alkyene-azide cycloaddition reaction system in >99% radiochemical conversions. Notably, the commonly used 18F-labeled azides were isolated through cartridges and directly used for cycloadditions, which greatly simplified the labeling procedure. The assembled triazolyl-tetrazines demonstrated high in vitro stability and reaction kinetics, exhibiting considerable potential for the development of PET agents.
Collapse
Affiliation(s)
- Cheng Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Frontiers Science Center for Disease Related Molecular Network West China Hospital, Sichuan University, Huaxi Research Building, 001 4th Keyuan Road, Chengdu 610041, China
| | - Kai Lu
- Department of Nuclear Medicine and Clinical Nuclear Medicine Research Lab, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jie Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Frontiers Science Center for Disease Related Molecular Network West China Hospital, Sichuan University, Huaxi Research Building, 001 4th Keyuan Road, Chengdu 610041, China
| | - Haoxing Wu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Frontiers Science Center for Disease Related Molecular Network West China Hospital, Sichuan University, Huaxi Research Building, 001 4th Keyuan Road, Chengdu 610041, China
| | - Wei Chen
- Department of Nuclear Medicine and Clinical Nuclear Medicine Research Lab, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
4
|
Leier S, Wuest F. Innovative Peptide Bioconjugation Chemistry with Radionuclides: Beyond Classical Click Chemistry. Pharmaceuticals (Basel) 2024; 17:1270. [PMID: 39458911 PMCID: PMC11510044 DOI: 10.3390/ph17101270] [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: 08/22/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Background: The incorporation of radionuclides into peptides and larger biomolecules requires efficient and sometimes biorthogonal reaction conditions, to which click chemistry provides a convenient approach. Methods: Traditionally, click-based radiolabeling techniques have focused on classical click chemistry, such as copper(I)-catalyzed alkyne-azide [3+2] cycloaddition (CuAAC), strain-promoted azide-alkyne [3+2] cycloaddition (SPAAC), traceless Staudinger ligation, and inverse electron demand Diels-Alder (IEDDA). Results: However, newly emerging click-based radiolabeling techniques, including tyrosine-click, sulfo-click, sulfur(VI) fluoride exchange (SuFEx), thiol-ene click, azo coupling, hydrazone formations, oxime formations, and RIKEN click offer valuable alternatives to classical click chemistry. Conclusions: This review will discuss the applications of these techniques in peptide radiochemistry.
Collapse
Affiliation(s)
- Samantha Leier
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Frank Wuest
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 1Z2, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2H1, Canada
- Department of Chemistry, Faculty of Science, University of Alberta, Edmonton, AB T6G 2G2, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB T6G 2R3, Canada
| |
Collapse
|
5
|
Adhikari K, Vanermen M, Da Silva G, Van den Wyngaert T, Augustyns K, Elvas F. Trans-cyclooctene-a Swiss army knife for bioorthogonal chemistry: exploring the synthesis, reactivity, and applications in biomedical breakthroughs. EJNMMI Radiopharm Chem 2024; 9:47. [PMID: 38844698 PMCID: PMC11156836 DOI: 10.1186/s41181-024-00275-x] [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: 03/25/2024] [Accepted: 05/27/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Trans-cyclooctenes (TCOs) are highly strained alkenes with remarkable reactivity towards tetrazines (Tzs) in inverse electron-demand Diels-Alder reactions. Since their discovery as bioorthogonal reaction partners, novel TCO derivatives have been developed to improve their reactivity, stability, and hydrophilicity, thus expanding their utility in diverse applications. MAIN BODY TCOs have garnered significant interest for their applications in biomedical settings. In chemical biology, TCOs serve as tools for bioconjugation, enabling the precise labeling and manipulation of biomolecules. Moreover, their role in nuclear medicine is substantial, with TCOs employed in the radiolabeling of peptides and other biomolecules. This has led to their utilization in pretargeted nuclear imaging and therapy, where they function as both bioorthogonal tags and radiotracers, facilitating targeted disease diagnosis and treatment. Beyond these applications, TCOs have been used in targeted cancer therapy through a "click-to-release" approach, in which they act as key components to selectively deliver therapeutic agents to cancer cells, thereby enhancing treatment efficacy while minimizing off-target effects. However, the search for a suitable TCO scaffold with an appropriate balance between stability and reactivity remains a challenge. CONCLUSIONS This review paper provides a comprehensive overview of the current state of knowledge regarding the synthesis of TCOs, and its challenges, and their development throughout the years. We describe their wide ranging applications as radiolabeled prosthetic groups for radiolabeling, as bioorthogonal tags for pretargeted imaging and therapy, and targeted drug delivery, with the aim of showcasing the versatility and potential of TCOs as valuable tools in advancing biomedical research and applications.
Collapse
Affiliation(s)
- Karuna Adhikari
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
- Molecular Imaging and Radiology, University of Antwerp, Antwerp, Belgium
| | - Maarten Vanermen
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
- Molecular Imaging and Radiology, University of Antwerp, Antwerp, Belgium
| | - Gustavo Da Silva
- Molecular Imaging and Radiology, University of Antwerp, Antwerp, Belgium
| | - Tim Van den Wyngaert
- Molecular Imaging and Radiology, University of Antwerp, Antwerp, Belgium
- Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Koen Augustyns
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium.
| | - Filipe Elvas
- Molecular Imaging and Radiology, University of Antwerp, Antwerp, Belgium.
- Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium.
| |
Collapse
|
6
|
Shalgunov V, van den Broek SL, Andersen IV, Raval NR, Schäfer G, Barz M, Herth MM, Battisti UM. Evaluation of F-537-Tetrazine in a model for brain pretargeting imaging. Comparison to N-(3-[ 18F] fluoro-5-(1,2,4,5-tetrazin-3-yl)benzyl)propan-1-amine. Nucl Med Biol 2024; 128-129:108877. [PMID: 38232579 DOI: 10.1016/j.nucmedbio.2024.108877] [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: 10/15/2023] [Revised: 01/05/2024] [Accepted: 01/05/2024] [Indexed: 01/19/2024]
Abstract
Brain pretargeted nuclear imaging for the diagnosis of various neurodegenerative diseases is a quickly developing field. The tetrazine ligation is currently the most explored approach to achieve this goal due to its remarkable properties. In this work, we evaluated the performance of F-537-Tetrazine, previously developed by Biogen, and N-(3-[18F]fluoro-5-(1,2,4,5-tetrazin-3-yl)benzyl)propan-1-amine, previously developed in our group, thereby allowing for the direct comparison of these two imaging probes. The evaluation included synthesis, radiolabeling and a comparison of the physicochemical properties of the compounds. Furthermore, their performance was evaluated by in vitro and in vivo pretargeting models. This study indicated that N-(3-[18F] fluoro-5-(1,2,4,5-tetrazin-3-yl)benzyl)propan-1-amine might be more suited for brain pretargeted imaging.
Collapse
Affiliation(s)
- Vladimir Shalgunov
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Sara Lopes van den Broek
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Ida Vang Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Nakul R Raval
- Neurobiology Research Unit and Center for Integrated Molecular Brain Imaging, Rigshospitalet Copenhagen University Hospital, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Gabriela Schäfer
- Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333CC Leiden, the Netherlands
| | - Matthias Barz
- Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333CC Leiden, the Netherlands
| | - Matthias M Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Umberto M Battisti
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| |
Collapse
|
7
|
Xu M, Ma X, Pigga JE, Zhang H, Wang S, Zhao W, Deng H, Wu AM, Liu R, Wu Z, Fox JM, Li Z. Development of 18F-Labeled hydrophilic trans-cyclooctene as a bioorthogonal tool for PET probe construction. Chem Commun (Camb) 2023; 59:14387-14390. [PMID: 37877355 PMCID: PMC10785124 DOI: 10.1039/d3cc04212j] [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: 10/26/2023]
Abstract
We report the development of a hydrophilic 18F-labeled a-TCO derivative [18F]3 (log P = 0.28) through a readily available precursor and a single-step radiofluorination reaction (RCY up to 52%). We demonstrated that [18F]3 can be used to construct not only multiple small molecule/peptide-based PET agents, but protein/diabody-based imaging probes in parallel.
Collapse
Affiliation(s)
- Muyun Xu
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA.
| | - Xinrui Ma
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA.
| | - Jessica E Pigga
- Department of Chemistry, the University of Delaware, Newark, Delaware, 19716, USA.
| | - He Zhang
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA.
| | - Shuli Wang
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA.
| | - Weiling Zhao
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA.
| | - Huaifu Deng
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA.
| | - Anna M Wu
- Department of Molecular Imaging and Therapy, Beckman Research Institute, City of Hope, Duarte, California, 91010, USA
| | - Rihe Liu
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Zhanhong Wu
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA.
| | - Joseph M Fox
- Department of Chemistry, the University of Delaware, Newark, Delaware, 19716, USA.
| | - Zibo Li
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA.
| |
Collapse
|
8
|
Panikar SS, Berry NK, Shmuel S, Keltee N, Pereira PM. In Vivo Biorthogonal Antibody Click for Dual Targeting and Augmented Efficacy in Cancer Treatment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.05.556426. [PMID: 37986985 PMCID: PMC10659283 DOI: 10.1101/2023.09.05.556426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Antibody-drug conjugates (ADCs) have emerged as promising therapeutics for cancer treatment; however, their effectiveness has been limited by single antigen targeting, potentially leading to resistance mechanisms triggered by tumor compensatory pathways or reduced expression of the target protein. Here, we present antibody-ADC click, an approach that harnesses bioorthogonal click chemistry for in vivo dual receptor targeting, irrespective of the levels of the tumor's expression of the ADC-targeting antigen. Antibody-ADC click enables targeting heterogeneity and enhances antibody internalization and drug delivery inside cancer cells, resulting in potent toxicity. We conjugated antibodies and ADCs to the bioorthogonal click moieties tetrazine (Tz) and trans-cyclooctene (TCO). Through sequential antibody administration in living biological systems, we achieved dual receptor targeting by in vivo clicking of antibody-TCO with antibody-Tz. We show that the clicked antibody therapy outperformed conventional ADC monotherapy or antibody combinations in preclinical models mimicking ADC-eligible, ADC-resistant, and ADC-ineligible tumors. Antibody-ADC click enables in vivo dual-antigen targeting without extensive antibody bioengineering, sustains tumor treatment, and enhances antibody-mediated cytotoxicity.
Collapse
Affiliation(s)
- Sandeep Surendra Panikar
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Na-Keysha Berry
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shayla Shmuel
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nai Keltee
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Patrícia M.R. Pereira
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
9
|
Halder R, Ma G, Rickmeier J, McDaniel JW, Petzold R, Neumann CN, Murphy JM, Ritter T. Deoxyfluorination of phenols for chemoselective 18F-labeling of peptides. Nat Protoc 2023; 18:3614-3651. [PMID: 37853158 DOI: 10.1038/s41596-023-00890-z] [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: 04/07/2023] [Accepted: 07/14/2023] [Indexed: 10/20/2023]
Abstract
The challenge of forming C-18F bonds is often a bottleneck in the development of new 18F-labeled tracer molecules for noninvasive functional imaging studies using positron emission tomography (PET). Nucleophilic aromatic substitution is the most widely employed reaction to functionalize aromatic substrates with the radioactive fluorine-18 but its scope is restricted to arenes containing electron-withdrawing substituents. Furthermore, many protic functional groups are incompatible with basic fluoride anions. Peptide substrates, which are highly desirable targets for PET molecular imaging, are particularly challenging to label with fluorine-18 because they are densely functionalized and sensitive to high temperatures and basic conditions. To expand the utility of nucleophilic aromatic substitution with fluorine-18, we describe two complementary procedures for the radiodeoxyfluorination of bench-stable and easy-to-access phenols that ensure rapid access to densely functionalized electron-rich and electron-poor 18F-aryl fluorides. The first procedure details the synthesis of an 18F-synthon and its subsequent ligation to the cysteine residue of Arg-Gly-Asp-Cys in 10.5 h from commercially available starting materials (189-min radiosynthesis). The second procedure describes the incorporation of commercially available CpRu(Fmoc-tyrosine)OTf into a fully protected peptide Lys-Met-Glu-(CpRu-Tyr)-Leu via solid-phase peptide synthesis and subsequent ruthenium-mediated uronium deoxyfluorination with fluorine-18 followed by deprotection, accomplished within 7 d (116-min radiosynthesis). Both radiolabeling methods are highly chemoselective and have conveniently been automated using commercially available radiosynthesis equipment so that the procedures described can be employed for the synthesis of peptide-based PET probes for in vivo imaging studies according to as low as reasonably achievable (ALARA) principles.
Collapse
Affiliation(s)
- Riya Halder
- Department of Organic Synthesis, Max-Planck-Institut für Kohlenforschung, Mülheim an der Ruhr, Germany
- Institute of Organic Chemistry, RWTH Aachen University, Aachen, Germany
| | - Gaoyuan Ma
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Strateos Inc., San Diego, CA, USA
| | - Jens Rickmeier
- Department of Organic Synthesis, Max-Planck-Institut für Kohlenforschung, Mülheim an der Ruhr, Germany
| | - James W McDaniel
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Roland Petzold
- Department of Organic Synthesis, Max-Planck-Institut für Kohlenforschung, Mülheim an der Ruhr, Germany
| | - Constanze N Neumann
- Department of Heterogeneous Catalysis, Max-Planck-Institut für Kohlenforschung, Mülheim an der Ruhr, Germany.
| | - 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, Los Angeles, CA, USA.
| | - Tobias Ritter
- Department of Organic Synthesis, Max-Planck-Institut für Kohlenforschung, Mülheim an der Ruhr, Germany.
| |
Collapse
|
10
|
Adhikari K, Dewulf J, Vangestel C, Van der Veken P, Stroobants S, Elvas F, Augustyns K. Characterization of Structurally Diverse 18F-Labeled d-TCO Derivatives as a PET Probe for Bioorthogonal Pretargeted Imaging. ACS OMEGA 2023; 8:38252-38262. [PMID: 37867688 PMCID: PMC10586181 DOI: 10.1021/acsomega.3c04597] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023]
Abstract
Background: The pretargeted imaging strategy using inverse electron demand Diels-Alder (IEDDA) cycloaddition between a trans-cyclooctene (TCO) and tetrazine (Tz) has emerged and rapidly grown as a promising concept to improve radionuclide imaging and therapy in oncology. This strategy has mostly relied on the use of radiolabeled Tz together with TCO-modified targeting vectors leading to a rapid growth of the number of available radiolabeled tetrazines, while only a few radiolabeled TCOs are currently reported. Here, we aim to develop novel and structurally diverse 18F-labeled cis-dioxolane-fused TCO (d-TCO) derivatives to further expand the bioorthogonal toolbox for in vivo ligation and evaluate their potential for positron emission tomography (PET) pretargeted imaging. Results: A small series of d-TCO derivatives were synthesized and tested for their reactivity against tetrazines, with all compounds showing fast reaction kinetics with tetrazines. A fluorescence-based pretargeted blocking study was developed to investigate the in vivo ligation of these compounds without labor-intensive prior radiochemical development. Two compounds showed excellent in vivo ligation results with blocking efficiencies of 95 and 97%. Two novel 18F-labeled d-TCO radiotracers were developed, from which [18F]MICA-214 showed good in vitro stability, favorable pharmacokinetics, and moderate in vivo stability. Micro-PET pretargeted imaging with [18F]MICA-214 in mice bearing LS174T tumors treated with tetrazine-modified CC49 monoclonal antibody (mAb) (CC49-Tz) showed significantly higher uptake in tumor tissue in the pretargeted group (CC49-Tz 2.16 ± 0.08% ID/mL) when compared to the control group with nonmodified mAb (CC49 1.34 ± 0.07% ID/mL). Conclusions: A diverse series of fast-reacting fluorinated d-TCOs were synthesized. A pretargeted blocking approach in tumor-bearing mice allowed the choice of a lead compound with fast reaction kinetics with Tz. A novel 18F-labeled d-TCO tracer was developed and used in a pretargeted PET imaging approach, allowing specific tumor visualization in a mouse model of colorectal cancer. Although further optimization of the radiotracer is needed to enhance the tumor-to-background ratios for pretargeted imaging, we anticipate that the 18F-labeled d-TCO will find use in studies where increased hydrophilicity and fast bioconjugation are required.
Collapse
Affiliation(s)
- Karuna Adhikari
- Laboratory
of Medicinal Chemistry, University of Antwerp, Antwerp 2610, Belgium
| | - Jonatan Dewulf
- Molecular
Imaging Center Antwerp, University of Antwerp, Antwerp 2610, Belgium
| | - Christel Vangestel
- Department
of Nuclear Medicine, Antwerp University
Hospital, Edegem 2650, Belgium
- Molecular
Imaging Center Antwerp, University of Antwerp, Antwerp 2610, Belgium
| | | | - Sigrid Stroobants
- Department
of Nuclear Medicine, Antwerp University
Hospital, Edegem 2650, Belgium
- Molecular
Imaging Center Antwerp, University of Antwerp, Antwerp 2610, Belgium
| | - Filipe Elvas
- Department
of Nuclear Medicine, Antwerp University
Hospital, Edegem 2650, Belgium
- Molecular
Imaging Center Antwerp, University of Antwerp, Antwerp 2610, Belgium
| | - Koen Augustyns
- Laboratory
of Medicinal Chemistry, University of Antwerp, Antwerp 2610, Belgium
| |
Collapse
|
11
|
Pakula RJ, Scott PJH. Applications of radiolabeled antibodies in neuroscience and neuro-oncology. J Labelled Comp Radiopharm 2023; 66:269-285. [PMID: 37322805 DOI: 10.1002/jlcr.4049] [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: 03/04/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/17/2023]
Abstract
Positron emission tomography (PET) is a powerful tool in medicine and drug development, allowing for non-invasive imaging and quantitation of biological processes in live organisms. Targets are often probed with small molecules, but antibody-based PET is expanding because of many benefits, including ease of design of new antibodies toward targets, as well as the very strong affinities that can be expected. Application of antibodies to PET imaging of targets in the central nervous system (CNS) is a particularly nascent field, but one with tremendous potential. In this review, we discuss the growth of PET in imaging of CNS targets, present the promises and progress in antibody-based CNS PET, explore challenges faced by the field, and discuss questions that this promising approach will need to answer moving forward for imaging and perhaps even radiotherapy.
Collapse
Affiliation(s)
- Ryan J Pakula
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Peter J H Scott
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
12
|
Rong J, Haider A, Jeppesen TE, Josephson L, Liang SH. Radiochemistry for positron emission tomography. Nat Commun 2023; 14:3257. [PMID: 37277339 PMCID: PMC10241151 DOI: 10.1038/s41467-023-36377-4] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 01/30/2023] [Indexed: 06/07/2023] Open
Abstract
Positron emission tomography (PET) constitutes a functional imaging technique that is harnessed to probe biological processes in vivo. PET imaging has been used to diagnose and monitor the progression of diseases, as well as to facilitate drug development efforts at both preclinical and clinical stages. The wide applications and rapid development of PET have ultimately led to an increasing demand for new methods in radiochemistry, with the aim to expand the scope of synthons amenable for radiolabeling. In this work, we provide an overview of commonly used chemical transformations for the syntheses of PET tracers in all aspects of radiochemistry, thereby highlighting recent breakthrough discoveries and contemporary challenges in the field. We discuss the use of biologicals for PET imaging and highlight general examples of successful probe discoveries for molecular imaging with PET - with a particular focus on translational and scalable radiochemistry concepts that have been entered to clinical use.
Collapse
Affiliation(s)
- Jian Rong
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA, 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Achi Haider
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA, 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Troels E Jeppesen
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Lee Josephson
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Steven H Liang
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA, 30322, USA.
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
13
|
Bauer D, Sarrett SM, Lewis JS, Zeglis BM. Click chemistry: a transformative technology in nuclear medicine. Nat Protoc 2023; 18:1659-1668. [PMID: 37100960 PMCID: PMC10293801 DOI: 10.1038/s41596-023-00825-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/10/2023] [Indexed: 04/28/2023]
Abstract
The 2022 Nobel Prize in Chemistry was awarded to Professors K. Barry Sharpless, Morten Meldal and Carolyn Bertozzi for their pioneering roles in the advent of click chemistry. Sharpless and Meldal worked to develop the canonical click reaction-the copper-catalyzed azide-alkyne cycloaddition-while Bertozzi opened new frontiers with the creation of the bioorthogonal strain-promoted azide-alkyne cycloaddition. These two reactions have revolutionized chemical and biological science by facilitating selective, high yielding, rapid and clean ligations and by providing unprecedented ways to manipulate living systems. Click chemistry has affected every aspect of chemistry and chemical biology, but few disciplines have been impacted as much as radiopharmaceutical chemistry. The importance of speed and selectivity in radiochemistry make it an almost tailor-made application of click chemistry. In this Perspective, we discuss the ways in which the copper-catalyzed azide-alkyne cycloaddition, the strain-promoted azide-alkyne cycloaddition and a handful of 'next-generation' click reactions have transformed radiopharmaceutical chemistry, both as tools for more efficient radiosyntheses and as linchpins of technologies that have the potential to improve nuclear medicine.
Collapse
Affiliation(s)
- David Bauer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samantha M Sarrett
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Chemistry, Hunter College of the City University of New York, New York, NY, USA
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY, USA
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA.
| | - Brian M Zeglis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Chemistry, Hunter College of the City University of New York, New York, NY, USA.
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY, USA.
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA.
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY, USA.
| |
Collapse
|
14
|
Lengacher R, Cosby AG, Śmiłowicz D, Boros E. Validation of a post-radiolabeling bioconjugation strategy for radioactive rare earth complexes with minimal structural footprint. Chem Commun (Camb) 2022; 58:13728-13730. [PMID: 36426996 PMCID: PMC9811989 DOI: 10.1039/d2cc06128g] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The nine-coordinate aza-macrocycle DO3Apic-NO2 and its kinetically inert rare earth complexes [M(DO3A-pic-NO2)]- (M = La, Tb, Eu, Lu, Y) can be readily bioconjugated to surface accessible thioles on peptides and proteins with a minimal structural footprint. All complexes express thioconjugation rate constants in the same order of magnitude (k = 0.3 h-1) with the exception of Sc (k = 0.89 h-1). Coupling to peptides and biologics with accessible cysteines also enables post-radiochelation bioconjugation at room temperature to afford injection-ready radiopharmaceuticals as demonstrated by formation of [177Lu][Lu(DO3Apic-NO2)]- and [86Y][Y(DO3Apic-NO2)]-, followed by post-labeling conjugation to a cysteine-functionalized peptide targeting the prostate specific membrane antigen. The 86Y-labeled construct efficiently localizes in target tumors with no significant off-target accumulation as evidenced by positron emission tomography, biodistribution studies and metabolite analysis.
Collapse
Affiliation(s)
- Raphael Lengacher
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
| | - Alexia G. Cosby
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
| | - Dariusz Śmiłowicz
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
| | - Eszter Boros
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
| |
Collapse
|
15
|
Antibody-Based In Vivo Imaging of Central Nervous System Targets-Evaluation of a Pretargeting Approach Utilizing a TCO-Conjugated Brain Shuttle Antibody and Radiolabeled Tetrazines. Pharmaceuticals (Basel) 2022; 15:ph15121445. [PMID: 36558900 PMCID: PMC9787164 DOI: 10.3390/ph15121445] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 11/09/2022] [Accepted: 11/15/2022] [Indexed: 11/25/2022] Open
Abstract
Bioorthogonal pretargeted imaging using the inverse-electron-demand Diels-Alder (IEDDA) reaction between a tetrazine (Tz) and a trans-cyclooctene (TCO) represents an attractive strategy for molecular imaging via antibodies. The advantages of using a pretargeted imaging approach are on the one hand the possibility to achieve a high signal-to-noise ratio and imaging contrast; on the other hand, the method allows the uncoupling of the biological half-life of antibodies from the physical half-life of short-lived radionuclides. A brain-penetrating antibody (mAb) specific for β-amyloid (Aβ) plaques was functionalized with TCO moieties for pretargeted labeling of Aβ plaques in vitro, ex vivo, and in vivo by a tritium-labeled Tz. The overall aim was to explore the applicability of mAbs for brain imaging, using a preclinical model system. In vitro clicked mAb-TCO-Tz was able to pass the blood-brain barrier of transgenic PS2APP mice and specifically visualize Aβ plaques ex vivo. Further experiments showed that click reactivity of the mAb-TCO construct in vivo persisted up to 3 days after injection by labeling Aβ plaques ex vivo after incubation of brain sections with the Tz in vitro. An attempted in vivo click reaction between injected mAb-TCO and Tz did not lead to significant labeling of Aβ plaques, most probably due to unfavorable in vivo properties of the used Tz and a long half-life of the mAb-TCO in the blood stream. This study clearly demonstrates that pretargeted imaging of CNS targets via antibody-based click chemistry is a viable approach. Further experiments are warranted to optimize the balance between stability and reactivity of all reactants, particularly the Tz.
Collapse
|
16
|
Li M, Ma X, Molnar CJ, Wang S, Wu Z, Popik VV, Li Z. Modular PET Agent Construction Strategy through Strain-Promoted Double-Click Reagent with Efficient Photoclick Step. Bioconjug Chem 2022; 33:2088-2096. [DOI: 10.1021/acs.bioconjchem.2c00427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Manshu Li
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Xinrui Ma
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Christopher J. Molnar
- Department of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Shuli Wang
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Zhanhong Wu
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Vladimir V. Popik
- Department of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Zibo Li
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
17
|
Liu L, Zhang D, Johnson M, Devaraj NK. Light-activated tetrazines enable precision live-cell bioorthogonal chemistry. Nat Chem 2022; 14:1078-1085. [PMID: 35788560 PMCID: PMC10198265 DOI: 10.1038/s41557-022-00963-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/03/2022] [Indexed: 02/07/2023]
Abstract
Bioorthogonal cycloaddition reactions between tetrazines and strained dienophiles are widely used for protein, lipid and glycan labelling because of their extremely rapid kinetics. However, controlling this chemistry in the presence of living mammalian cells with a high degree of spatial and temporal precision remains a challenge. Here we demonstrate a versatile approach to light-activated formation of tetrazines from photocaged dihydrotetrazines. Photouncaging, followed by spontaneous transformation to reactive tetrazine, enables live-cell spatiotemporal control of rapid bioorthogonal cycloaddition with dienophiles such as trans-cyclooctenes. Photocaged dihydrotetrazines are stable in conditions that normally degrade tetrazines, enabling efficient early-stage incorporation of bioorthogonal handles into biomolecules such as peptides. Photocaged dihydrotetrazines allow the use of non-toxic light to trigger tetrazine ligations on living mammalian cells. By tagging reactive phospholipids with fluorophores, we demonstrate modification of HeLa cell membranes with single-cell spatial resolution. Finally, we show that photo-triggered therapy is possible by coupling tetrazine photoactivation with strategies that release prodrugs in response to tetrazine ligation.
Collapse
Affiliation(s)
- Luping Liu
- Department of Chemistry and Biochemistry, University of California, San Diego, CA, USA
| | - Dongyang Zhang
- Department of Chemistry and Biochemistry, University of California, San Diego, CA, USA
| | - Mai Johnson
- Department of Chemistry and Biochemistry, University of California, San Diego, CA, USA
| | - Neal K Devaraj
- Department of Chemistry and Biochemistry, University of California, San Diego, CA, USA.
| |
Collapse
|
18
|
Recent Advances in the Development of Tetrazine Ligation Tools for Pretargeted Nuclear Imaging. Pharmaceuticals (Basel) 2022; 15:ph15060685. [PMID: 35745604 PMCID: PMC9227058 DOI: 10.3390/ph15060685] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 01/25/2023] Open
Abstract
Tetrazine ligation has gained interest as a bio-orthogonal chemistry tool within the last decade. In nuclear medicine, tetrazine ligation is currently being explored for pretargeted approaches, which have the potential to revolutionize state-of-the-art theranostic strategies. Pretargeting has been shown to increase target-to-background ratios for radiopharmaceuticals based on nanomedicines, especially within early timeframes. This allows the use of radionuclides with short half-lives which are more suited for clinical applications. Pretargeting bears the potential to increase the therapeutic dose delivered to the target as well as reduce the respective dose to healthy tissue. Combined with the possibility to be applied for diagnostic imaging, pretargeting could be optimal for theranostic approaches. In this review, we highlight efforts that have been made to radiolabel tetrazines with an emphasis on imaging.
Collapse
|
19
|
Yang E, Liu Q, Huang G, Liu J, Wei W. Engineering nanobodies for next-generation molecular imaging. Drug Discov Today 2022; 27:1622-1638. [PMID: 35331925 DOI: 10.1016/j.drudis.2022.03.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 02/04/2022] [Accepted: 03/17/2022] [Indexed: 12/12/2022]
Abstract
In recent years, nanobodies have emerged as ideal imaging agents for molecular imaging. Molecular nanobody imaging combines the specificity of nanobodies with the sensitivity of state-of-the-art molecular imaging modalities, such as positron emission tomography (PET). Given that modifications of nanobodies alter their pharmacokinetics (PK), the engineering strategies that combine nanobodies with radionuclides determine the effectiveness, reliability, and safety of the molecular imaging probes. In this review, we introduce conjugation strategies that have been applied to nanobodies, including random conjugation, 99mTc tricarbonyl chemistry, sortase A-mediated site-specific conjugation, maleimide-cysteine chemistry, and click chemistries. We also summarize the latest advances in nanobody tracers, emphasizing their preclinical and clinical use. In addition, we elaborate on nanobody-based near-infrared fluorescence (NIRF) imaging and image-guided surgery.
Collapse
Affiliation(s)
- Erpeng Yang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Qiufang Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China.
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China.
| |
Collapse
|
20
|
Battisti UM, García-Vázquez R, Svatunek D, Herrmann B, Löffler A, Mikula H, Herth MM. Synergistic Experimental and Computational Investigation of the Bioorthogonal Reactivity of Substituted Aryltetrazines. Bioconjug Chem 2022; 33:608-624. [PMID: 35290735 PMCID: PMC9026259 DOI: 10.1021/acs.bioconjchem.2c00042] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
![]()
Tetrazines (Tz) have
been applied as bioorthogonal agents for various
biomedical applications, including pretargeted imaging approaches.
In radioimmunoimaging, pretargeting increases the target-to-background
ratio while simultaneously reducing the radiation burden. We have
recently reported a strategy to directly 18F-label highly
reactive tetrazines based on a 3-(3-fluorophenyl)-Tz core structure.
Herein, we report a kinetic study on this versatile scaffold. A library
of 40 different tetrazines was prepared, fully characterized, and
investigated with an emphasis on second-order rate constants for the
reaction with trans-cyclooctene (TCO). Our results
reveal the effects of various substitution patterns and moreover demonstrate
the importance of measuring reactivities in the solvent of interest,
as click rates in different solvents do not necessarily correlate
well. In particular, we report that tetrazines modified in the 2-position
of the phenyl substituent show high intrinsic reactivity toward TCO,
which is diminished in aqueous systems by unfavorable solvent effects.
The obtained results enable the prediction of the bioorthogonal reactivity
and thereby facilitate the development of the next generation of substituted
aryltetrazines for in vivo applications.
Collapse
Affiliation(s)
- Umberto M Battisti
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark
| | - Rocío García-Vázquez
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark.,Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Dennis Svatunek
- Institute of Applied Synthetic Chemistry, Technische Universität Wien (TU Wien), Getreidemarkt 9, 1060 Vienna, Austria
| | - Barbara Herrmann
- Institute of Applied Synthetic Chemistry, Technische Universität Wien (TU Wien), Getreidemarkt 9, 1060 Vienna, Austria
| | - Andreas Löffler
- Institute of Applied Synthetic Chemistry, Technische Universität Wien (TU Wien), Getreidemarkt 9, 1060 Vienna, Austria
| | - Hannes Mikula
- Institute of Applied Synthetic Chemistry, Technische Universität Wien (TU Wien), Getreidemarkt 9, 1060 Vienna, Austria
| | - Matthias Manfred Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark.,Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| |
Collapse
|
21
|
García-Vázquez R, Jørgensen JT, Bratteby KE, Shalgunov V, Hvass L, Herth MM, Kjær A, Battisti UM. Development of 18F-Labeled Bispyridyl Tetrazines for In Vivo Pretargeted PET Imaging. Pharmaceuticals (Basel) 2022; 15:ph15020245. [PMID: 35215356 PMCID: PMC8879724 DOI: 10.3390/ph15020245] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 02/05/2023] Open
Abstract
Pretargeted PET imaging is an emerging and fast-developing method to monitor immuno-oncology strategies. Currently, tetrazine ligation is considered the most promising bioorthogonal reaction for pretargeting in vivo. Recently, we have developed a method to 18F-label ultrareactive tetrazines by copper-mediated fluorinations. However, bispyridyl tetrazines—one of the most promising structures for in vivo pretargeted applications—were inaccessible using this strategy. We believed that our successful efforts to 18F-label H-tetrazines using low basic labeling conditions could also be used to label bispyridyl tetrazines via aliphatic nucleophilic substitution. Here, we report the first direct 18F-labeling of bispyridyl tetrazines, their optimization for in vivo use, as well as their successful application in pretargeted PET imaging. This strategy resulted in the design of [18F]45, which could be labeled in a satisfactorily radiochemical yield (RCY = 16%), molar activity (Am = 57 GBq/µmol), and high radiochemical purity (RCP > 98%). The [18F]45 displayed a target-to-background ratio comparable to previously successfully applied tracers for pretargeted imaging. This study showed that bispyridyl tetrazines can be developed into pretargeted imaging agents. These structures allow an easy chemical modification of 18F-labeled tetrazines, paving the road toward highly functionalized pretargeting tools. Moreover, bispyridyl tetrazines led to near-instant drug release of iTCO-tetrazine-based ‘click-to-release’ reactions. Consequently, 18F-labeled bispyridyl tetrazines bear the possibility to quantify such release in vivo in the future.
Collapse
Affiliation(s)
- Rocío García-Vázquez
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; (R.G.-V.); (K.E.B.); (V.S.)
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Jesper Tranekjær Jørgensen
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen, Denmark; (J.T.J.); (L.H.)
| | - Klas Erik Bratteby
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; (R.G.-V.); (K.E.B.); (V.S.)
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
- Department of Radiation Physics, Skåne University Hospital, Barngatan 3, 22242 Lund, Sweden
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; (R.G.-V.); (K.E.B.); (V.S.)
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Lars Hvass
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen, Denmark; (J.T.J.); (L.H.)
| | - Matthias M. Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; (R.G.-V.); (K.E.B.); (V.S.)
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
- Correspondence: (M.M.H.); (A.K.); (U.M.B.)
| | - Andreas Kjær
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen, Denmark; (J.T.J.); (L.H.)
- Correspondence: (M.M.H.); (A.K.); (U.M.B.)
| | - Umberto Maria Battisti
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; (R.G.-V.); (K.E.B.); (V.S.)
- Correspondence: (M.M.H.); (A.K.); (U.M.B.)
| |
Collapse
|
22
|
Zhang C, Yan K, Fu C, Peng H, Hawker CJ, Whittaker AK. Biological Utility of Fluorinated Compounds: from Materials Design to Molecular Imaging, Therapeutics and Environmental Remediation. Chem Rev 2022; 122:167-208. [PMID: 34609131 DOI: 10.1021/acs.chemrev.1c00632] [Citation(s) in RCA: 176] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The applications of fluorinated molecules in bioengineering and nanotechnology are expanding rapidly with the controlled introduction of fluorine being broadly studied due to the unique properties of C-F bonds. This review will focus on the design and utility of C-F containing materials in imaging, therapeutics, and environmental applications with a central theme being the importance of controlling fluorine-fluorine interactions and understanding how such interactions impact biological behavior. Low natural abundance of fluorine is shown to provide sensitivity and background advantages for imaging and detection of a variety of diseases with 19F magnetic resonance imaging, 18F positron emission tomography and ultrasound discussed as illustrative examples. The presence of C-F bonds can also be used to tailor membrane permeability and pharmacokinetic properties of drugs and delivery agents for enhanced cell uptake and therapeutics. A key message of this review is that while the promise of C-F containing materials is significant, a subset of highly fluorinated compounds such as per- and polyfluoroalkyl substances (PFAS), have been identified as posing a potential risk to human health. The unique properties of the C-F bond and the significant potential for fluorine-fluorine interactions in PFAS structures necessitate the development of new strategies for facile and efficient environmental removal and remediation. Recent progress in the development of fluorine-containing compounds as molecular imaging and therapeutic agents will be reviewed and their design features contrasted with environmental and health risks for PFAS systems. Finally, present challenges and future directions in the exploitation of the biological aspects of fluorinated systems will be described.
Collapse
Affiliation(s)
- Cheng Zhang
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Queensland, Brisbane, Queensland 4072, Australia
- Materials Research Laboratory, University of California, Santa Barbara, California 93106, United States
| | - Kai Yan
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
- National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science and Technology, Xi'an 710021, China
- Xi'an Key Laboratory of Green Chemicals and Functional Materials, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Changkui Fu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Hui Peng
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Craig J Hawker
- Materials Research Laboratory, University of California, Santa Barbara, California 93106, United States
- Materials Department, University of California, Santa Barbara, California 93106, United States
- Department of Chemistry & Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Andrew K Whittaker
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
23
|
Battisti UM, Bratteby K, Jørgensen JT, Hvass L, Shalgunov V, Mikula H, Kjær A, Herth MM. Development of the First Aliphatic 18F-Labeled Tetrazine Suitable for Pretargeted PET Imaging-Expanding the Bioorthogonal Tool Box. J Med Chem 2021; 64:15297-15312. [PMID: 34649424 DOI: 10.1021/acs.jmedchem.1c01326] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Pretargeted imaging of nanomedicines have attracted considerable interest because it has the potential to increase imaging contrast while reducing radiation burden to healthy tissue. Currently, the tetrazine ligation is the fastest bioorthogonal reaction for this strategy and, consequently, the state-of-art choice for in vivo chemistry. We have recently identified key properties for tetrazines in pretargeting. We have also developed a method to 18F-label reactive tetrazines using an aliphatic nucleophilic substitution strategy. Here, we combined this knowledge and developed an 18F-labeled tetrazine for pretargeted imaging. In order to develop this ligand, a small SAR study was performed. The most promising compound was selected for labeling and subsequent positron-emission-tomography in vivo imaging. Radiolabeling was achieved in satisfactory yields, molar activities, and high radiochemical purities. [18F]15 displayed favorable pharmacokinetics and remarkable target-to-background ratios-as early as 1 h post injection. We believe that this agent could be a promising candidate for translation into clinical studies.
Collapse
Affiliation(s)
- Umberto M Battisti
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Klas Bratteby
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.,Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark.,Department of Radiation Physics, Skåne University Hospital, Barngatan 3, 22242 Lund, Sweden
| | - Jesper T Jørgensen
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen Ø, Denmark
| | - Lars Hvass
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen Ø, Denmark
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Hannes Mikula
- Institute of Applied Synthetic Chemistry, Technische Universität Wien (TU Wien), Getreidemarkt 9, 1060 Vienna, Austria
| | - Andreas Kjær
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen Ø, Denmark
| | - Matthias Manfred Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.,Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| |
Collapse
|
24
|
Bratteby K, Shalgunov V, Battisti UM, Petersen IN, van den Broek SL, Ohlsson T, Gillings N, Erlandsson M, Herth MM. Insights into Elution of Anion Exchange Cartridges: Opening the Path toward Aliphatic 18F-Radiolabeling of Base-Sensitive Tracers. ACS Pharmacol Transl Sci 2021; 4:1556-1566. [PMID: 34661074 PMCID: PMC8506604 DOI: 10.1021/acsptsci.1c00133] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Indexed: 01/16/2023]
Abstract
![]()
Aliphatic nucleophilic
substitution (SN2) with [18F]fluoride is the
most widely applied method to prepare 18F-labeled positron
emission tomography (PET) tracers. Strong
basic conditions commonly used during 18F-labeling procedures
inherently limit or prohibit labeling of base-sensitive scaffolds.
The high basicity stems from the tradition to trap [18F]fluoride
on anion exchange cartridges and elute it afterward with basic anions.
This sequence is used to facilitate the transfer of [18F]fluoride from an aqueous to an aprotic organic, polar reaction
medium, which is beneficial for SN2 reactions. Furthermore,
this sequence also removes cationic radioactive contaminations from
cyclotron-irradiated [18O]water from which [18F]fluoride is produced. In this study, we developed an efficient
elution procedure resulting in low basicity that permits SN2 18F-labeling of base-sensitive scaffolds. Extensive
screening of trapping and elution conditions (>1000 experiments)
and
studying their influence on the radiochemical yield (RCY) allowed
us to identify a suitable procedure for this. Using this procedure,
four PET tracers and three synthons could be radiolabeled in substantially
higher RCYs (up to 2.5-fold) compared to those of previously published
procedures, even from lower precursor amounts. Encouraged by these
results, we applied our low-basicity method to the radiolabeling of
highly base-sensitive tetrazines, which cannot be labeled using state-of-art
direct aliphatic 18F-labeling procedures. Labeling succeeded
in RCYs of up to 20%. We believe that our findings facilitate PET
tracer development by opening the path toward simple and direct SN2 18F fluorination of base-sensitive substrates.
Collapse
Affiliation(s)
- Klas Bratteby
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark.,Department of Radiation Physics, Skåne University Hospital, Barngatan 3, 22242 Lund, Sweden.,Department of Clinical Physiology Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark.,Department of Clinical Physiology Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Umberto Maria Battisti
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark
| | - Ida Nyman Petersen
- Department of Clinical Physiology Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Sara Lopes van den Broek
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark
| | - Tomas Ohlsson
- Department of Radiation Physics, Skåne University Hospital, Barngatan 3, 22242 Lund, Sweden
| | - Nic Gillings
- Department of Clinical Physiology Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Maria Erlandsson
- Department of Radiation Physics, Skåne University Hospital, Barngatan 3, 22242 Lund, Sweden
| | - Matthias M Herth
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark.,Department of Clinical Physiology Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| |
Collapse
|
25
|
Biedka S, Schmidt BF, Frey NM, Boothman SM, Minden JS, Lucas A. Reversible Click Chemistry Tag for Universal Proteome Sample Preparation for Top-Down and Bottom-Up Analysis. J Proteome Res 2021; 20:4787-4800. [PMID: 34524823 DOI: 10.1021/acs.jproteome.1c00443] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Successful proteome analysis requires reliable sample preparation beginning with protein solubilization and ending with a sample free of contaminants, ready for downstream analysis. Most proteome sample preparation technologies utilize precipitation or filter-based separation, both of which have significant disadvantages. None of the current technologies are able to prepare both intact proteins or digested peptides. Here, we introduce a reversible protein tag, ProMTag, that enables whole proteome capture, cleanup, and release of intact proteins for top-down analysis. Alternatively, the addition of a novel Trypsin derivative to the workflow generates peptides for bottom-up analysis. We show that the ProMTag workflow yields >90% for intact proteins and >85% for proteome digests. For top-down analysis, ProMTag cleanup improves resolution on 2D gels; for bottom-up exploration, this methodology produced reproducible mass spectrometry results, demonstrating that the ProMTag method is a truly universal approach that produces high-quality proteome samples compatible with multiple downstream analytical techniques. Data are available via ProteomeXchange with identifier PXD027799.
Collapse
Affiliation(s)
- Stephanie Biedka
- Impact Proteomics, LLC., Pittsburgh, Pennsylvania 15206, United States
| | - Brigitte F Schmidt
- JGS Research Co., Pittsburgh, Pennsylvania 15212, United States.,Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Nolan M Frey
- Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Sarah M Boothman
- Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Jonathan S Minden
- Impact Proteomics, LLC., Pittsburgh, Pennsylvania 15206, United States.,Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Amber Lucas
- Impact Proteomics, LLC., Pittsburgh, Pennsylvania 15206, United States
| |
Collapse
|
26
|
García-Vázquez R, Battisti UM, Jørgensen JT, Shalgunov V, Hvass L, Stares DL, Petersen IN, Crestey F, Löffler A, Svatunek D, Kristensen JL, Mikula H, Kjaer A, Herth MM. Direct Cu-mediated aromatic 18F-labeling of highly reactive tetrazines for pretargeted bioorthogonal PET imaging. Chem Sci 2021; 12:11668-11675. [PMID: 34659701 PMCID: PMC8442695 DOI: 10.1039/d1sc02789a] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/25/2021] [Indexed: 11/21/2022] Open
Abstract
Pretargeted imaging can be used to visualize and quantify slow-accumulating targeting vectors with short-lived radionuclides such as fluorine-18 - the most popular clinically applied Positron Emission Tomography (PET) radionuclide. Pretargeting results in higher target-to-background ratios compared to conventional imaging approaches using long-lived radionuclides. Currently, the tetrazine ligation is the most popular bioorthogonal reaction for pretargeted imaging, but a direct 18F-labeling strategy for highly reactive tetrazines, which would be highly beneficial if not essential for clinical translation, has thus far not been reported. In this work, a simple, scalable and reliable direct 18F-labeling procedure has been developed. We initially studied the applicability of different leaving groups and labeling methods to develop this procedure. The copper-mediated 18F-labeling exploiting stannane precursors showed the most promising results. This approach was then successfully applied to a set of tetrazines, including highly reactive H-tetrazines, suitable for pretargeted PET imaging. The labeling succeeded in radiochemical yields (RCYs) of up to approx. 25%. The new procedure was then applied to develop a pretargeting tetrazine-based imaging agent. The tracer was synthesized in a satisfactory RCY of ca. 10%, with a molar activity of 134 ± 22 GBq μmol-1 and a radiochemical purity of >99%. Further evaluation showed that the tracer displayed favorable characteristics (target-to-background ratios and clearance) that may qualify it for future clinical translation.
Collapse
Affiliation(s)
- Rocío García-Vázquez
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen Jagtvej 160 2100 Copenhagen Denmark
- Department of Clinical Physiology, Nuclear Medicine & PET Rigshospitalet, Blegdamsvej 9 2100 Copenhagen Denmark
| | - Umberto M Battisti
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen Jagtvej 160 2100 Copenhagen Denmark
| | - Jesper T Jørgensen
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen Blegdamsvej 9 2100 Copenhagen Denmark
- Department of Clinical Physiology, Nuclear Medicine & PET Rigshospitalet, Blegdamsvej 9 2100 Copenhagen Denmark
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen Jagtvej 160 2100 Copenhagen Denmark
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen Blegdamsvej 9 2100 Copenhagen Denmark
- Department of Clinical Physiology, Nuclear Medicine & PET Rigshospitalet, Blegdamsvej 9 2100 Copenhagen Denmark
| | - Lars Hvass
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen Blegdamsvej 9 2100 Copenhagen Denmark
- Department of Clinical Physiology, Nuclear Medicine & PET Rigshospitalet, Blegdamsvej 9 2100 Copenhagen Denmark
| | - Daniel L Stares
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen Jagtvej 160 2100 Copenhagen Denmark
| | - Ida N Petersen
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen Blegdamsvej 9 2100 Copenhagen Denmark
| | - François Crestey
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen Jagtvej 160 2100 Copenhagen Denmark
| | - Andreas Löffler
- Institute of Applied Synthetic Chemistry, Technische Universität Wien (TU Wien) Getreidemarkt 9 1060 Vienna Austria
| | - Dennis Svatunek
- Institute of Applied Synthetic Chemistry, Technische Universität Wien (TU Wien) Getreidemarkt 9 1060 Vienna Austria
| | - Jesper L Kristensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen Jagtvej 160 2100 Copenhagen Denmark
| | - Hannes Mikula
- Institute of Applied Synthetic Chemistry, Technische Universität Wien (TU Wien) Getreidemarkt 9 1060 Vienna Austria
| | - Andreas Kjaer
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen Blegdamsvej 9 2100 Copenhagen Denmark
- Department of Clinical Physiology, Nuclear Medicine & PET Rigshospitalet, Blegdamsvej 9 2100 Copenhagen Denmark
| | - Matthias M Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen Jagtvej 160 2100 Copenhagen Denmark
- Department of Clinical Physiology, Nuclear Medicine & PET Rigshospitalet, Blegdamsvej 9 2100 Copenhagen Denmark
| |
Collapse
|
27
|
Cheewawisuttichai T, Brichacek M. Development of a multifunctional neoglycoside auxiliary for applications in glycomics research. Org Biomol Chem 2021; 19:6613-6617. [PMID: 34264248 DOI: 10.1039/d1ob00941a] [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: 11/21/2022]
Abstract
A novel, multifunctional, tetrazine-containing neoglycoside auxiliary has been synthesized in three steps and 28% overall yield. The oxyamine was conjugated with unprotected carbohydrates under aqueous conditions (pH = 4.7), with DMF as a cosolvent, to provide neoglycosides in yields ranging between 51% and 68%. This auxiliary displayed broad advantages in the isolation and purification of complex carbohydrate mixtures, compatibility during extension by glycosyltransferases, and direct conjugation to chemical probes. Furthermore, the auxiliary can be removed in 96% yield under acidic conditions (0.25% TFA in H2O) that leave glycosidic linkages intact. Thereby, the tetrazine-containing neoglycoside auxiliary can serve to facilitate future glycomics investigations.
Collapse
|
28
|
Bratteby K, Shalgunov V, Herth MM. Aliphatic 18 F-Radiofluorination: Recent Advances in the Labeling of Base-Sensitive Substrates*. ChemMedChem 2021; 16:2612-2622. [PMID: 34169672 DOI: 10.1002/cmdc.202100303] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Indexed: 12/19/2022]
Abstract
Aliphatic fluorine-18 radiolabeling is the most commonly used method to synthesize tracers for PET-imaging. With an increasing demand for 18 F-radiotracers for clinical applications, new labeling strategies aiming to increase radiochemical yields of established tracers or, more importantly, to enable 18 F-labeling of new scaffolds have been developed. In recent years, increased attention has been focused on the direct aliphatic 18 F-fluorination of base-sensitive substrates in this respect. This minireview gives a concise overview of the recent advances within this field and aims to highlight the advantages and limitations of these methods.
Collapse
Affiliation(s)
- Klas Bratteby
- Department of Drug Design and Pharmacology Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100, Copenhagen, Denmark.,Department of Radiation Physics, Skåne University Hospital, Barngatan 3, 222 42, Lund, Sweden.,Department of Clinical Physiology, Nuclear Medicine & PET Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100, Copenhagen, Denmark
| | - Matthias Manfred Herth
- Department of Drug Design and Pharmacology Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100, Copenhagen, Denmark.,Department of Clinical Physiology, Nuclear Medicine & PET Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| |
Collapse
|
29
|
Handula M, Chen KT, Seimbille Y. IEDDA: An Attractive Bioorthogonal Reaction for Biomedical Applications. Molecules 2021; 26:molecules26154640. [PMID: 34361793 PMCID: PMC8347371 DOI: 10.3390/molecules26154640] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/26/2022] Open
Abstract
The pretargeting strategy has recently emerged in order to overcome the limitations of direct targeting, mainly in the field of radioimmunotherapy (RIT). This strategy is directly dependent on chemical reactions, namely bioorthogonal reactions, which have been developed for their ability to occur under physiological conditions. The Staudinger ligation, the copper catalyzed azide-alkyne cycloaddition (CuAAC) and the strain-promoted [3 + 2] azide–alkyne cycloaddition (SPAAC) were the first bioorthogonal reactions introduced in the literature. However, due to their incomplete biocompatibility and slow kinetics, the inverse-electron demand Diels-Alder (IEDDA) reaction was advanced in 2008 by Blackman et al. as an optimal bioorthogonal reaction. The IEDDA is the fastest bioorthogonal reaction known so far. Its biocompatibility and ideal kinetics are very appealing for pretargeting applications. The use of a trans-cyclooctene (TCO) and a tetrazine (Tz) in the reaction encouraged researchers to study them deeply. It was found that both reagents are sensitive to acidic or basic conditions. Furthermore, TCO is photosensitive and can be isomerized to its cis-conformation via a radical catalyzed reaction. Unfortunately, the cis-conformer is significantly less reactive toward tetrazine than the trans-conformation. Therefore, extensive research has been carried out to optimize both click reagents and to employ the IEDDA bioorthogonal reaction in biomedical applications.
Collapse
Affiliation(s)
- Maryana Handula
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands;
| | - Kuo-Ting Chen
- Department of Chemistry, National Dong Hwa University, Shoufeng, Hualien 974301, Taiwan;
| | - Yann Seimbille
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands;
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Correspondence: ; Tel.: +31-10-703-8961
| |
Collapse
|
30
|
Fu Y, Helbert H, Simeth NA, Crespi S, Spoelstra GB, van Dijl JM, van Oosten M, Nazario LR, van der Born D, Luurtsema G, Szymanski W, Elsinga PH, Feringa BL. Ultrafast Photoclick Reaction for Selective 18F-Positron Emission Tomography Tracer Synthesis in Flow. J Am Chem Soc 2021; 143:10041-10047. [PMID: 34181410 PMCID: PMC8283755 DOI: 10.1021/jacs.1c02229] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
![]()
The development of
very fast, clean, and selective methods for
indirect labeling in PET tracer synthesis is an ongoing challenge.
Here we present the development of an ultrafast photoclick method
for the synthesis of short-lived 18F-PET tracers based
on the photocycloaddition reaction of 9,10-phenanthrenequinones
with electron-rich alkenes. The respective precursors are synthetically
easily accessible and can be functionalized with various target groups.
Using a flow photo-microreactor, the photoclick reaction can be performed
in 60 s, and clinically relevant tracers for prostate cancer and bacterial
infection imaging were prepared to demonstrate practicality of the
method.
Collapse
Affiliation(s)
- Youxin Fu
- Centre for Systems Chemistry, Stratingh Institute for Chemistry, Faculty for Science and Engineering, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Hugo Helbert
- Centre for Systems Chemistry, Stratingh Institute for Chemistry, Faculty for Science and Engineering, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands.,Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Nadja A Simeth
- Centre for Systems Chemistry, Stratingh Institute for Chemistry, Faculty for Science and Engineering, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Stefano Crespi
- Centre for Systems Chemistry, Stratingh Institute for Chemistry, Faculty for Science and Engineering, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Gerbren B Spoelstra
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Jan Maarten van Dijl
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Marleen van Oosten
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Luiza Reali Nazario
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Dion van der Born
- FutureChemistry, Agro Business Park 10, 6708 PW Wageningen, The Netherlands
| | - Gert Luurtsema
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Wiktor Szymanski
- Centre for Systems Chemistry, Stratingh Institute for Chemistry, Faculty for Science and Engineering, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands.,Department of Radiology, Medical Imaging Center, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Philip H Elsinga
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Ben L Feringa
- Centre for Systems Chemistry, Stratingh Institute for Chemistry, Faculty for Science and Engineering, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| |
Collapse
|
31
|
Allott L, Amgheib A, Barnes C, Braga M, Brickute D, Wang N, Fu R, Ghaem-Maghami S, Aboagye EO. Radiolabelling an 18F biologic via facile IEDDA "click" chemistry on the GE FASTLab™ platform. REACT CHEM ENG 2021; 6:1070-1078. [PMID: 34123410 PMCID: PMC8167423 DOI: 10.1039/d1re00117e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023]
Abstract
The use of biologics in positron emission tomography (PET) imaging is an important area of radiopharmaceutical development and new automated methods are required to facilitate their production. We report an automated radiosynthesis method to produce a radiolabelled biologic via facile inverse electron demand Diels-Alder (IEDDA) "click" chemistry on a single GE FASTLab™ cassette. We exemplified the method by producing a fluorine-18 radiolabelled interleukin-2 (IL2) radioconjugate from a trans-cyclooctene (TCO) modified IL2 precursor. The radioconjugate was produced using a fully automated radiosynthesis on a single FASTLab™ cassette in a decay-corrected radiochemical yield (RCY, d.c.) of 19.8 ± 2.6% in 110 min (from start of synthesis); the molar activity was 132.3 ± 14.6 GBq μmol-1. The in vitro uptake of [18F]TTCO-IL2 correlated with the differential receptor expression (CD25, CD122, CD132) in PC3, NK-92 and activated human PBMCs. The automated method may be adapted for the radiosynthesis of any TCO-modified protein via IEDDA chemistry.
Collapse
Affiliation(s)
- Louis Allott
- Comprehensive Cancer Imaging Centre, Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
- Positron Emission Tomography Research Centre, Faculty of Health Sciences, University of Hull Cottingham Road Kingston upon Hull HU6 7RX UK
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Hull Cottingham Road Kingston upon Hull HU6 7RX UK
| | - Ala Amgheib
- Comprehensive Cancer Imaging Centre, Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
- Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
| | - Chris Barnes
- Comprehensive Cancer Imaging Centre, Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
| | - Marta Braga
- Comprehensive Cancer Imaging Centre, Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
| | - Diana Brickute
- Comprehensive Cancer Imaging Centre, Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
| | - Ning Wang
- Comprehensive Cancer Imaging Centre, Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
| | - Ruisi Fu
- Comprehensive Cancer Imaging Centre, Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
| | - Sadaf Ghaem-Maghami
- Comprehensive Cancer Imaging Centre, Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
- Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
| | - Eric O Aboagye
- Comprehensive Cancer Imaging Centre, Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
| |
Collapse
|
32
|
Stéen EJ, Jørgensen JT, Denk C, Battisti UM, Nørregaard K, Edem PE, Bratteby K, Shalgunov V, Wilkovitsch M, Svatunek D, Poulie CBM, Hvass L, Simón M, Wanek T, Rossin R, Robillard M, Kristensen JL, Mikula H, Kjaer A, Herth MM. Lipophilicity and Click Reactivity Determine the Performance of Bioorthogonal Tetrazine Tools in Pretargeted In Vivo Chemistry. ACS Pharmacol Transl Sci 2021; 4:824-833. [PMID: 33860205 PMCID: PMC8033778 DOI: 10.1021/acsptsci.1c00007] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Indexed: 12/12/2022]
Abstract
The development of highly selective and fast biocompatible reactions for ligation and cleavage has paved the way for new diagnostic and therapeutic applications of pretargeted in vivo chemistry. The concept of bioorthogonal pretargeting has attracted considerable interest, in particular for the targeted delivery of radionuclides and drugs. In nuclear medicine, pretargeting can provide increased target-to-background ratios at early time-points compared to traditional approaches. This reduces the radiation burden to healthy tissue and, depending on the selected radionuclide, enables better imaging contrast or higher therapeutic efficiency. Moreover, bioorthogonally triggered cleavage of pretargeted antibody-drug conjugates represents an emerging strategy to achieve controlled release and locally increased drug concentrations. The toolbox of bioorthogonal reactions has significantly expanded in the past decade, with the tetrazine ligation being the fastest and one of the most versatile in vivo chemistries. Progress in the field, however, relies heavily on the development and evaluation of (radio)labeled compounds, preventing the use of compound libraries for systematic studies. The rational design of tetrazine probes and triggers has thus been impeded by the limited understanding of the impact of structural parameters on the in vivo ligation performance. In this work, we describe the development of a pretargeted blocking assay that allows for the investigation of the in vivo fate of a structurally diverse library of 45 unlabeled tetrazines and their capability to reach and react with pretargeted trans-cyclooctene (TCO)-modified antibodies in tumor-bearing mice. This study enabled us to assess the correlation of click reactivity and lipophilicity of tetrazines with their in vivo performance. In particular, high rate constants (>50 000 M-1 s-1) for the reaction with TCO and low calculated logD 7.4 values (below -3) of the tetrazine were identified as strong indicators for successful pretargeting. Radiolabeling gave access to a set of selected 18F-labeled tetrazines, including highly reactive scaffolds, which were used in pretargeted PET imaging studies to confirm the results from the blocking study. These insights thus enable the rational design of tetrazine probes for in vivo application and will thereby assist the clinical translation of bioorthogonal pretargeting.
Collapse
Affiliation(s)
- E. Johanna
L. Stéen
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
- Department
of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej
9, 2100 Copenhagen, Denmark
| | - Jesper T. Jørgensen
- Department
of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej
9, 2100 Copenhagen, Denmark
- Cluster
for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen Ø, Denmark
| | - Christoph Denk
- Institute
of Applied Synthetic Chemistry, Technische
Universität Wien (TU Wien), Getreidemarkt 9, 1060 Vienna, Austria
| | - Umberto M. Battisti
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Kamilla Nørregaard
- Department
of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej
9, 2100 Copenhagen, Denmark
- Cluster
for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen Ø, Denmark
| | - Patricia E. Edem
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
- Cluster
for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen Ø, Denmark
| | - Klas Bratteby
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
- Department
of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej
9, 2100 Copenhagen, Denmark
- Department
of Radiation Physics, Skåne University
Hospital, Barngatan 3, 22242 Lund, Sweden
| | - Vladimir Shalgunov
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
- Cluster
for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen Ø, Denmark
| | - Martin Wilkovitsch
- Institute
of Applied Synthetic Chemistry, Technische
Universität Wien (TU Wien), Getreidemarkt 9, 1060 Vienna, Austria
| | - Dennis Svatunek
- Institute
of Applied Synthetic Chemistry, Technische
Universität Wien (TU Wien), Getreidemarkt 9, 1060 Vienna, Austria
| | - Christian B. M. Poulie
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
- Cluster
for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen Ø, Denmark
| | - Lars Hvass
- Department
of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej
9, 2100 Copenhagen, Denmark
- Cluster
for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen Ø, Denmark
| | - Marina Simón
- Department
of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej
9, 2100 Copenhagen, Denmark
- Cluster
for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen Ø, Denmark
| | - Thomas Wanek
- Preclinical
Molecular Imaging, AIT Austrian Institute
of Technology GmbH, 2444 Seibersdorf, Austria
| | - Raffaella Rossin
- Tagworks
Pharmaceuticals, Geert
Grooteplein 10, 6525 GA Nijmegen, Netherlands
| | - Marc Robillard
- Tagworks
Pharmaceuticals, Geert
Grooteplein 10, 6525 GA Nijmegen, Netherlands
| | - Jesper L. Kristensen
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Hannes Mikula
- Institute
of Applied Synthetic Chemistry, Technische
Universität Wien (TU Wien), Getreidemarkt 9, 1060 Vienna, Austria
| | - Andreas Kjaer
- Department
of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej
9, 2100 Copenhagen, Denmark
- Cluster
for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen Ø, Denmark
| | - Matthias M. Herth
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
- Department
of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej
9, 2100 Copenhagen, Denmark
| |
Collapse
|
33
|
Wilkovitsch M, Haider M, Sohr B, Herrmann B, Klubnick J, Weissleder R, Carlson JCT, Mikula H. A Cleavable C 2-Symmetric trans-Cyclooctene Enables Fast and Complete Bioorthogonal Disassembly of Molecular Probes. J Am Chem Soc 2020; 142:19132-19141. [PMID: 33119297 PMCID: PMC7662912 DOI: 10.1021/jacs.0c07922] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Indexed: 12/15/2022]
Abstract
Bioorthogonal chemistry is bridging the divide between static chemical connectivity and the dynamic physiologic regulation of molecular state, enabling in situ transformations that drive multiple technologies. In spite of maturing mechanistic understanding and new bioorthogonal bond-cleavage reactions, the broader goal of molecular ON/OFF control has been limited by the inability of existing systems to achieve both fast (i.e., seconds to minutes, not hours) and complete (i.e., >99%) cleavage. To attain the stringent performance characteristics needed for high fidelity molecular inactivation, we have designed and synthesized a new C2-symmetric trans-cyclooctene linker (C2TCO) that exhibits excellent biological stability and can be rapidly and completely cleaved with functionalized alkyl-, aryl-, and H-tetrazines, irrespective of click orientation. By incorporation of C2TCO into fluorescent molecular probes, we demonstrate highly efficient extracellular and intracellular bioorthogonal disassembly via omnidirectional tetrazine-triggered cleavage.
Collapse
Affiliation(s)
- Martin Wilkovitsch
- Institute
of Applied Synthetic Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria
| | - Maximilian Haider
- Institute
of Applied Synthetic Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria
| | - Barbara Sohr
- Institute
of Applied Synthetic Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria
| | - Barbara Herrmann
- Institute
of Applied Synthetic Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria
| | - Jenna Klubnick
- Center
for Systems Biology, Massachusetts General
Hospital Research Institute, Boston, Massachusetts 02114, United States
| | - Ralph Weissleder
- Center
for Systems Biology, Massachusetts General
Hospital Research Institute, Boston, Massachusetts 02114, United States
- Department
of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jonathan C. T. Carlson
- Center
for Systems Biology, Massachusetts General
Hospital Research Institute, Boston, Massachusetts 02114, United States
- Cancer
Center, Massachusetts General Hospital and
Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Hannes Mikula
- Institute
of Applied Synthetic Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria
| |
Collapse
|
34
|
Zhang T, Cai J, Wang H, Wang M, Yuan H, Wu Z, Ma X, Li Z. RXH-Reactive 18F-Vinyl Sulfones as Versatile Agents for PET Probe Construction. Bioconjug Chem 2020; 31:2482-2487. [PMID: 33103415 DOI: 10.1021/acs.bioconjchem.0c00487] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Efficient radiolabeling reactions are important chemical tools in biomedical research especially in probe construction. Herein, three 18F-labeled vinyl sulfones were prepared. In particular, 18F-PEG1-VS (((2-(2-(fluoro-18F)ethoxy)ethyl)sulfonyl)ethane) could not only allow chemoselective labeling of bioactive molecules containing -XH (X = S, NH) groups, but also react with red blood cells both in vitro and in living mice for potential cell tracking applications. In addition, these hydrophilic agents were found to cross the blood brain barrier (BBB) efficiently and localize at the cerebellum region. In summary, 18F-labeled vinyl sulfones provide a versatile platform for PET probe construction.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, 125 Mason Farm Road, Chapel Hill, North Carolina 27599, United States
| | - Jianhua Cai
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, 125 Mason Farm Road, Chapel Hill, North Carolina 27599, United States.,School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, P. R. China
| | - Hui Wang
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, 125 Mason Farm Road, Chapel Hill, North Carolina 27599, United States
| | - Mengzhe Wang
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, 125 Mason Farm Road, Chapel Hill, North Carolina 27599, United States
| | - Hong Yuan
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, 125 Mason Farm Road, Chapel Hill, North Carolina 27599, United States
| | - Zhanhong Wu
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, 125 Mason Farm Road, Chapel Hill, North Carolina 27599, United States
| | - Xiaofen Ma
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, 125 Mason Farm Road, Chapel Hill, North Carolina 27599, United States.,Department of Medical Imaging, Guangdong Second Provincial General Hospital. 466 Xingang Middle Road, Haizhu District, Guangzhou City, Guangdong Province 510317, P. R. China
| | - Zibo Li
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, 125 Mason Farm Road, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
35
|
Zhou Z, Zalutsky MR, Vaidyanathan G. Labeling a TCO-functionalized single domain antibody fragment with 18F via inverse electron demand Diels Alder cycloaddition using a fluoronicotinyl moiety-bearing tetrazine derivative. Bioorg Med Chem 2020; 28:115634. [PMID: 32773089 DOI: 10.1016/j.bmc.2020.115634] [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: 03/13/2020] [Revised: 06/27/2020] [Accepted: 06/28/2020] [Indexed: 02/07/2023]
Abstract
Single domain antibody fragments (sdAbs) exhibit a rapid tumor uptake and fast blood clearance amenable for labeling with 18F (t½ = 110 min) but suffer from high kidney accumulation. Previously, we developed a method for 18F-labeling of sdAbs via trans-cyclooctene (TCO)-tetrazine (Tz) inverse electron demand Diel's Alder cycloaddition reaction (IEDDAR) that incorporated a renal brush border enzyme (RBBE)-cleavable linker. Although >15 fold reduction in kidney activity levels was achieved, tumor uptake was compromised. Here we investigate whether replacing the [18F]AlF-NOTA moiety with [18F]fluoronicotinyl would rectify this problem. Anti-HER2 sdAb 5F7 was first derivatized with a TCO-containing agent that included the RBBE-cleavable linker GlyLys (GK) and a PEG chain, and then subjected to IEDDAR with 6-[18F]fluoronicotinyl-PEG4-methyltetrazine to provide [18F]FN-PEG4-Tz-TCO-GK-PEG4-5F7 ([18F]FN-GK-5F7). For comparisons, a control lacking GK linker and 5F7 labeled using residualizing N-succinimidyl 3-guanidinomethyl-5-[125I]iodobenzoate (iso-[125I]SGMIB) also were synthesized. Radiochemical purity, affinity (KD) and immunoreactive fraction of [18F]FN-GK-5F7 were 99%, 5.4 ± 0.7 nM and 72.5 ± 4.3%, respectively. Tumor uptake of [18F]FN-GK-5F7 in athymic mice bearing subcutaneous SKOV3 xenografts (3.7 ± 1.2% ID/g and 3.4 ± 1.0% ID/g at 1 h and 3 h, respectively) was 2- to 3-fold lower than for co-injected iso-[125I]SGMIB-5F7 (6.9 ± 1.9 %ID/g and 8.7 ± 3.0 %ID/g). However, due to its 6-fold lower kidney activity levels, tumor-to-kidney ratios for [18F]FN-GK-5F7 were 3-4 times higher than those for co-injected iso-[125I]SGMIB-5F7 as well as those observed for the 18F conjugate lacking the RBBE-cleavable linker. Micro-PET/CT imaging of [18F]FN-GK-5F7 in mice with SKOV-3 subcutaneous xenografts clearly delineated tumor as early as 1 h with minimal activity in the kidneys; however, there was considerable activity in gallbladder and intestines. Although the tumor uptake of [18F]FN-GK-5F7 was unexpectedly disappointing, incorporating an alternative RBBE-cleavable linker into this labeling strategy may ameliorate this problem.
Collapse
Affiliation(s)
- Zhengyuan Zhou
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Michael R Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
36
|
Longo B, Zanato C, Piras M, Dall'Angelo S, Windhorst AD, Vugts DJ, Baldassarre M, Zanda M. Design, Synthesis, Conjugation, and Reactivity of Novel trans,trans-1,5-Cyclooctadiene-Derived Bioorthogonal Linkers. Bioconjug Chem 2020; 31:2201-2210. [PMID: 32786505 DOI: 10.1021/acs.bioconjchem.0c00375] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The tetrazine/trans-cyclooctene (TCO) inverse electron-demand Diels-Alder (IEDDA) reaction is the fastest bioorthogonal "click" ligation process reported to date. In this context, TCO reagents have found widespread applications; however, their availability and structural diversity is still somewhat limited due to challenges connected with their synthesis and structural modification. To address this issue, we developed a novel strategy for the conjugation of TCO derivatives to a biomolecule, which allows for the creation of greater structural diversity from a single precursor molecule, i.e., trans,trans-1,5-cyclooctadiene [(E,E)-COD] 1, whose preparation requires standard laboratory equipment and readily available reagents. This two-step strategy relies on the use of new bifunctional TCO linkers (5a-11a) for IEDDA reactions, which can be synthesized via 1,3-dipolar cycloaddition of (E,E)-COD 1 with different azido spacers (5-11) carrying an electrophilic function (NHS-ester, N-succinimidyl carbonate, p-nitrophenyl-carbonate, maleimide) in the ω-position. Following bioconjugation of these electrophilic linkers to the nucleophilic residue (cysteine or lysine) of a protein (step 1), the resulting TCO-decorated constructs can be subjected to a IEDDA reaction with tetrazines functionalized with fluorescent or near-infrared (NIR) tags (step 2). We successfully used this strategy to label bovine serum albumin with the TCO linker 8a and subsequently reacted it in a cell lysate with the fluorescein-isothiocyanate (FITC)-derived tetrazine 12. The same strategy was then used to label the bacterial wall of Gram-positive Staphylococcus aureus, showing the potential of these linkers for live-cell imaging. Finally, we determined the impact of structural differences of the linkers upon the stability of the bioorthogonal constructs. The compounds for stability studies were prepared by conjugation of TCO linkers 6a, 8a, and 10a to mAbs, such as Rituximab and Obinutuzumab, and subsequent labeling with a reactive Cy3-functionalized tetrazine.
Collapse
Affiliation(s)
- Beatrice Longo
- Institute of Medical Sciences, Foresterhill, University of Aberdeen, AB252ZD Aberdeen, United Kingdom.,Centre for Sensing and Imaging Science, School of Science, Loughborough University, LB11 3TU Loughborough, United Kingdom
| | - Chiara Zanato
- Institute of Medical Sciences, Foresterhill, University of Aberdeen, AB252ZD Aberdeen, United Kingdom
| | - Monica Piras
- Institute of Medical Sciences, Foresterhill, University of Aberdeen, AB252ZD Aberdeen, United Kingdom
| | - Sergio Dall'Angelo
- Institute of Medical Sciences, Foresterhill, University of Aberdeen, AB252ZD Aberdeen, United Kingdom
| | - Albert D Windhorst
- Amsterdam UMC, Vrije Universiteit, dept. Radiology and Nuclear Medicine, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Danielle J Vugts
- Amsterdam UMC, Vrije Universiteit, dept. Radiology and Nuclear Medicine, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Massimiliano Baldassarre
- Institute of Medical Sciences, Foresterhill, University of Aberdeen, AB252ZD Aberdeen, United Kingdom
| | - Matteo Zanda
- Institute of Medical Sciences, Foresterhill, University of Aberdeen, AB252ZD Aberdeen, United Kingdom.,Centre for Sensing and Imaging Science, School of Science, Loughborough University, LB11 3TU Loughborough, United Kingdom.,CNR-SCITEC, via Mancinelli 7, 20131 Milan, Italy
| |
Collapse
|
37
|
Dewulf J, Adhikari K, Vangestel C, Wyngaert TVD, Elvas F. Development of Antibody Immuno-PET/SPECT Radiopharmaceuticals for Imaging of Oncological Disorders-An Update. Cancers (Basel) 2020; 12:E1868. [PMID: 32664521 PMCID: PMC7408676 DOI: 10.3390/cancers12071868] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 01/12/2023] Open
Abstract
Positron emission tomography (PET) and single-photon emission computed tomography (SPECT) are molecular imaging strategies that typically use radioactively labeled ligands to selectively visualize molecular targets. The nanomolar sensitivity of PET and SPECT combined with the high specificity and affinity of monoclonal antibodies have shown great potential in oncology imaging. Over the past decades a wide range of radio-isotopes have been developed into immuno-SPECT/PET imaging agents, made possible by novel conjugation strategies (e.g., site-specific labeling, click chemistry) and optimization and development of novel radiochemistry procedures. In addition, new strategies such as pretargeting and the use of antibody fragments have entered the field of immuno-PET/SPECT expanding the range of imaging applications. Non-invasive imaging techniques revealing tumor antigen biodistribution, expression and heterogeneity have the potential to contribute to disease diagnosis, therapy selection, patient stratification and therapy response prediction achieving personalized treatments for each patient and therefore assisting in clinical decision making.
Collapse
Affiliation(s)
- Jonatan Dewulf
- Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium; (J.D.); (C.V.); (T.V.D.W.)
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, B-2650 Edegem, Belgium
| | - Karuna Adhikari
- Faculty of Pharmaceutical Biomedical and Veterinary Sciences, Medicinal Chemistry, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium;
| | - Christel Vangestel
- Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium; (J.D.); (C.V.); (T.V.D.W.)
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, B-2650 Edegem, Belgium
| | - Tim Van Den Wyngaert
- Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium; (J.D.); (C.V.); (T.V.D.W.)
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, B-2650 Edegem, Belgium
| | - Filipe Elvas
- Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium; (J.D.); (C.V.); (T.V.D.W.)
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, B-2650 Edegem, Belgium
| |
Collapse
|
38
|
Affiliation(s)
- Matteo Zanda
- Loughborough University Centre for Sensing and Imaging Science School of Science Sir David Davies Building, Chemistry Department 113TU Loughborough United Kingdom
- C.N.R.‐SCITEC Via Mancinelli 7 20131 Milano Italy
| | - Raffaella Bucci
- Loughborough University Centre for Sensing and Imaging Science School of Science Sir David Davies Building, Chemistry Department 113TU Loughborough United Kingdom
| | - Nikki L. Sloan
- Loughborough University Centre for Sensing and Imaging Science School of Science Sir David Davies Building, Chemistry Department 113TU Loughborough United Kingdom
| | - Lydia Topping
- Loughborough University Centre for Sensing and Imaging Science School of Science Sir David Davies Building, Chemistry Department 113TU Loughborough United Kingdom
| |
Collapse
|
39
|
Chen Z, Chen M, Zhou K, Rao J. Pre‐targeted Imaging of Protease Activity through In Situ Assembly of Nanoparticles. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201916352] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Zixin Chen
- Departments of Radiology and Chemistry Molecular Imaging Program at Stanford Stanford University School of Medicine Stanford CA 94305 USA
| | - Min Chen
- Departments of Radiology and Chemistry Molecular Imaging Program at Stanford Stanford University School of Medicine Stanford CA 94305 USA
| | - Kaixiang Zhou
- Departments of Radiology and Chemistry Molecular Imaging Program at Stanford Stanford University School of Medicine Stanford CA 94305 USA
| | - Jianghong Rao
- Departments of Radiology and Chemistry Molecular Imaging Program at Stanford Stanford University School of Medicine Stanford CA 94305 USA
| |
Collapse
|
40
|
Chen Z, Chen M, Zhou K, Rao J. Pre-targeted Imaging of Protease Activity through In Situ Assembly of Nanoparticles. Angew Chem Int Ed Engl 2020; 59:7864-7870. [PMID: 32056345 DOI: 10.1002/anie.201916352] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/11/2020] [Indexed: 02/06/2023]
Abstract
The pre-targeted imaging of enzyme activity has not been reported, likely owing to the lack of a mechanism to retain the injected substrate in the first step for subsequent labeling. Herein, we report the use of two bioorthogonal reactions-the condensation reaction of aromatic nitriles and aminothiols and the inverse-electron demand Diels-Alder reaction between tetrazine and trans-cyclooctene (TCO)-to develop a novel strategy for pre-targeted imaging of the activity of proteases. The substrate probe (TCO-C-SNAT4) can be selectively activated by an enzyme target (e.g. caspase-3/7), which triggers macrocyclization and subsequent in situ self-assembly into nanoaggregates retained at the target site. The tetrazine-imaging tag conjugate labels TCO in the nanoaggregates to generate selective signal retention for imaging in vitro, in cells, and in mice. Owing to the decoupling of enzyme activation and imaging tag immobilization, TCO-C-SNAT4 can be repeatedly injected to generate and accumulate more TCO-nanoaggregates for click labeling.
Collapse
Affiliation(s)
- Zixin Chen
- Departments of Radiology and Chemistry, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Min Chen
- Departments of Radiology and Chemistry, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Kaixiang Zhou
- Departments of Radiology and Chemistry, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jianghong Rao
- Departments of Radiology and Chemistry, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
41
|
Feng H, Zhang H, Wang M, Vannam R, Wang H, Yan X, Ouyang W, Jia X, Fox JM, Li Z. Improving Tumor-to-Background Contrast through Hydrophilic Tetrazines: The Construction of 18 F-Labeled PET Agents Targeting Nonsmall Cell Lung Carcinoma. Chemistry 2020; 26:4690-4694. [PMID: 32030822 DOI: 10.1002/chem.202000028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/04/2020] [Indexed: 02/04/2023]
Abstract
Bioorthogonal reactions have been widely used in the biomedical field. 18 F-TCO/Tetrazine ligation is the most reactive radiolabelled inverse electron demand Diels-Alder reaction, but its application had been limited due to modest contrast ratios of the resulting conjugates. Herein, we describe the use of hydrophilic tetrazines to improve tumor-to-background contrast of neurotensin receptor targeted PET agents. PET agents were constructed using a rapid Diels-Alder reaction of the radiolabeled trans-cyclooctene (18 F-sTCO) with neurotensin (NT) conjugates of a 3,6-diaryltetrazine, 3-methyl-6-aryltetrazine, and a derivative of 3,6-di(2-hydroxyethyl)tetrazine. Although cell binding assays demonstrated all agents have comparable binding affinity, the conjugate derived from 3,6-di(2-hydroxyethyl)tetrazine demonstrated the highest tumor to muscle contrast, followed by conjugates of the 3-methyl-6-aryltetrazine and 3,6-diaryltetrazine.
Collapse
Affiliation(s)
- Huijuan Feng
- Department of Nuclear Medicine, ZhuJiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510280, P. R. China.,Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA
| | - He Zhang
- Department of Material Science and Engineering, University of Delaware, Newark, Delaware, 19716, USA
| | - Mengzhe Wang
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA
| | - Raghu Vannam
- Brown Laboratories, Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware, 19716, USA
| | - Hui Wang
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA
| | - Xuefeng Yan
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA
| | - Wei Ouyang
- Department of Nuclear Medicine, ZhuJiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510280, P. R. China
| | - Xinqiao Jia
- Department of Material Science and Engineering, University of Delaware, Newark, Delaware, 19716, USA
| | - Joseph M Fox
- Brown Laboratories, Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware, 19716, USA
| | - Zibo Li
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA
| |
Collapse
|
42
|
Evaluation of Organo [ 18F]Fluorosilicon Tetrazine as a Prosthetic Group for the Synthesis of PET Radiotracers. Molecules 2020; 25:molecules25051208. [PMID: 32156020 PMCID: PMC7179430 DOI: 10.3390/molecules25051208] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 01/09/2023] Open
Abstract
Fluorine-18 is the most widely used positron emission tomography (PET) radionuclide currently in clinical application, due to its optimal nuclear properties. The synthesis of 18F-labeled radiotracers often requires harsh reaction conditions, limiting the use of sensitive bio- and macromolecules as precursors for direct radiolabeling with fluorine-18. We aimed to develop a milder and efficient in vitro and in vivo labeling method for trans-cyclooctene (TCO) functionalized proteins, through the bioorthogonal inverse-electron demand Diels-Alder (IEDDA) reaction with fluorine-18 radiolabeled tetrazine ([18F]SiFA-Tz). Here, we used TCO-modified bovine serum albumin (BSA) as the model protein, and isotopic exchange (IE) (19F/18F) chemistry as the labeling strategy. The radiolabeling of albumin-TCO with [18F]SiFA-Tz ([18F]6), providing [18F]fluoroalbumin ([18F]10) in high radiochemical yield (99.1 ± 0.2%, n = 3) and a molar activity (MA) of 1.1 GBq/µmol, confirmed the applicability of [18F]6 as a quick in vitro fluorination reagent for the TCO functionalized proteins. While the biological evaluation of [18F]6 demonstrated defluorination in vivo, limiting the utility for pretargeted applications, the in vivo stability of the radiotracer was dramatically improved when [18F]6 was used for the radiolabeling of albumin-TCO ([18F]10) in vitro, prior to administration. Due to the detected defluorination in vivo, structural optimization of the prosthetic group for improved stability is needed before further biological studies and application of pretargeted PET imaging.
Collapse
|
43
|
Gamache RF, Zettlitz KA, Tsai WTK, Collins J, Wu AM, Murphy JM. Tri-functional platform for construction of modular antibody fragments for in vivo 18F-PET or NIRF molecular imaging. Chem Sci 2020; 11:1832-1838. [PMID: 34123276 PMCID: PMC8148382 DOI: 10.1039/c9sc05007h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Positron emission tomography (PET) molecular imaging is a powerful tool for interrogating physiological and biochemical processes to understand the biology of disease and advance therapeutic developments. Near-infrared fluorescence (NIRF) optical imaging has become increasingly popular for intraoperative staging to enable cellular resolution imaging of tumor margins during surgical resection. In addition, engineered antibody fragments have emerged as promising molecular imaging agents given their exquisite target selectivity, rapid systemic clearance and site-selective chemical modification. We report a tri-functional platform for construction of a modular antibody fragment that can rapidly be labeled with radionuclides or fluorophores for PET or NIRF molecular imaging of prostate stem cell antigen (PSCA). To provide a universal approach towards the targeted delivery of PET and optical imaging agents, we have developed a tri-functional platform (TFP) for the facile construction of modular, target-specific tracers.![]()
Collapse
Affiliation(s)
- Raymond F Gamache
- Department of Chemistry and Biochemistry, University of California Los Angeles CA 90095 USA
| | - Kirstin A Zettlitz
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California Los Angeles CA 90095 USA
| | - Wen-Ting K Tsai
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California Los Angeles CA 90095 USA
| | - Jeffrey Collins
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California Los Angeles CA 90095 USA
| | - Anna M Wu
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California Los Angeles CA 90095 USA
| | - 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 CA 90095 USA
| |
Collapse
|
44
|
Park SH, Seo JY, Park JY, Ji YB, Kim K, Choi HS, Choi S, Kim JH, Min BH, Kim MS. An injectable, click-crosslinked, cytomodulin-modified hyaluronic acid hydrogel for cartilage tissue engineering. NPG ASIA MATERIALS 2019; 11:30. [DOI: 10.1038/s41427-019-0130-1] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/26/2019] [Accepted: 03/31/2019] [Indexed: 01/05/2025]
Abstract
AbstractThis is the first report, to our knowledge, of the preparation of an injectable in situ–forming click-crosslinked hyaluronic acid (Cx-HA) hydrogel (Cx-HA-CM) containing chemical immobilized cytomodulin-2 (CM), a chondrogenic differentiation factor, and on the utility of human periodontal ligament stem cells (hPLSCs) as a cell source for cartilage tissue engineering. hPLSCs served here as a stem cell source tolerant to ex vivo manipulation. CM induced in vitro chondrogenic differentiation of hPLSCs comparable to induction with traditional TGF-β. Cx-HA was prepared via a click-reaction between tetrazine-modified HA and transcyclooctene-modified HA. Cx-HA displayed significantly more features of a stiff hydrogel than HA. Cx-HA had a three-dimensional porous interconnected structure, absorbed a large volume of biological medium, and showed excellent biocompatibility. In contrast to HA, the Cx-HA hydrogel persisted in vitro and in vivo for an extended period, as evidenced by in vivo near-infrared fluorescence imaging. CM covalently linked to Cx-HA (Cx-HA-CM) remained inside Cx-HA for a prolonged period compared with CM physically loaded onto Cx-HA [Cx-HA (+CM)]. Cx-HA-CM also caused better chondrogenic differentiation of hPLSCs, as evidenced by Alcian blue and Safranin O staining, and greater increases in the expression of type II collagen, glycosaminoglycan content and SOX9, aggrecan, and type 2α1 collagen mRNA levels. Thus, compared to Cx-HA (+CM), the hPLSC-loaded Cx-HA-CM hydrogel induced greater chondrogenic differentiation of hPLSCs via CM that was retained in the hydrogel for a much longer period of time.
Collapse
|
45
|
|
46
|
Neumann S, Biewend M, Rana S, Binder WH. The CuAAC: Principles, Homogeneous and Heterogeneous Catalysts, and Novel Developments and Applications. Macromol Rapid Commun 2019; 41:e1900359. [PMID: 31631449 DOI: 10.1002/marc.201900359] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/06/2019] [Indexed: 01/08/2023]
Abstract
The copper-catalyzed azide/alkyne cycloaddition reaction (CuAAC) has emerged as the most useful "click" chemistry. Polymer science has profited enormously from CuAAC by its simplicity, ease, scope, applicability and efficiency. Basic principles of the CuAAC are reviewed with a focus on homogeneous and heterogeneous catalysts, ligands, anchimeric assistance, and basic chemical principles. Recent developments of ligand design and acceleration are discussed.
Collapse
Affiliation(s)
- Steve Neumann
- Institute of Chemistry, Chair of Macromolecular Chemistry, Martin-Luther University Halle-Wittenberg, von Danckelmannplatz 4, D-06120, Halle (Saale), Germany
| | - Michel Biewend
- Institute of Chemistry, Chair of Macromolecular Chemistry, Martin-Luther University Halle-Wittenberg, von Danckelmannplatz 4, D-06120, Halle (Saale), Germany
| | - Sravendra Rana
- School of Engineering University of Petroleum and Energy Studies (UPES), Dehradun, Uttarakhand, 248007, India
| | - Wolfgang H Binder
- Institute of Chemistry, Chair of Macromolecular Chemistry, Martin-Luther University Halle-Wittenberg, von Danckelmannplatz 4, D-06120, Halle (Saale), Germany
| |
Collapse
|
47
|
Recent Advances in Bioorthogonal Click Chemistry for Efficient Synthesis of Radiotracers and Radiopharmaceuticals. Molecules 2019; 24:molecules24193567. [PMID: 31581645 PMCID: PMC6803924 DOI: 10.3390/molecules24193567] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/23/2019] [Accepted: 09/27/2019] [Indexed: 12/17/2022] Open
Abstract
In recent years, several catalyst-free site-specific reactions have been investigated for the efficient conjugation of biomolecules, nanomaterials, and living cells. Representative functional group pairs for these reactions include the following: (1) azide and cyclooctyne for strain-promoted cycloaddition reaction, (2) tetrazine and trans-alkene for inverse-electron-demand-Diels–Alder reaction, and (3) electrophilic heterocycles and cysteine for rapid condensation/addition reaction. Due to their excellent specificities and high reaction rates, these conjugation methods have been utilized for the labeling of radioisotopes (e.g., radiohalogens, radiometals) to various target molecules. The radiolabeled products prepared by these methods have been applied to preclinical research, such as in vivo molecular imaging, pharmacokinetic studies, and radiation therapy of cancer cells. In this review, we explain the basics of these chemical reactions and introduce their recent applications in the field of radiopharmacy and chemical biology. In addition, we discuss the significance, current challenges, and prospects of using bioorthogonal conjugation reactions.
Collapse
|
48
|
Wu ZC, Boger DL. Synthesis, Characterization, and Cycloaddition Reactivity of a Monocyclic Aromatic 1,2,3,5-Tetrazine. J Am Chem Soc 2019; 141:16388-16397. [PMID: 31524389 DOI: 10.1021/jacs.9b07744] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Herein we disclose the synthesis and full characterization of the first monocyclic aromatic 1,2,3,5-tetrazine, 4,6-diphenyl-1,2,3,5-tetrazine. Initial studies of its cycloaddition reactivity, mode, regioselectivity, and scope illustrate that it participates as the 4π-component of well-behaved inverse electron demand Diels-Alder reactions where it preferentially reacts with electron-rich or strained dienophiles. It was found to exhibit an intrinsic reactivity comparable to that of the isomeric 3,6-diphenyl-1,2,4,5-tetrazine, display a single mode of cycloaddition with reaction only across C4/N1 (no N2/N5 cycloaddition observed), proceed with a predictable regioselectivity (dienophile most electron-rich atom attaches to C4), and manifest additional reactivity complementary to the isomeric 1,2,4,5-tetrazines. It not only exhibits a remarkable cycloaddition reactivity, surprisingly good stability (e.g., stable to chromatography, long-term storage, presence of H2O even as reaction co-solvent), and broad cycloaddition scope, but it also displays powerful orthogonal reactivity with the 1,2,4,5-tetrazines. Whereas the latter reacts at extraordinary cycloaddition rates with strained dienophiles (tetrazine ligation), the new and isomeric 1,2,3,5-tetrazine displays similarly remarkable cycloaddition rates and efficiencies with amidines (1,2,3,5-tetrazine/amidine ligation). The crossover reactivities (1,2,4,5-tetrazines with amidines and 1,2,3,5-tetrazines with strained dienophiles) are sufficiently low to indicate they may be capable of use concurrently without competitive reactions.
Collapse
Affiliation(s)
- Zhi-Chen Wu
- Department of Chemistry and The Skaggs Institute for Chemical Biology , The Scripps Research Institute , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - Dale L Boger
- Department of Chemistry and The Skaggs Institute for Chemical Biology , The Scripps Research Institute , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| |
Collapse
|
49
|
Dong P, Wang X, Zheng J, Zhang X, Li Y, Wu H, Li L. Recent Advances in Targeting Nuclear Molecular Imaging Driven by Tetrazine Bioorthogonal Chemistry. Curr Med Chem 2019; 27:3924-3943. [PMID: 31267851 DOI: 10.2174/1386207322666190702105829] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 04/18/2019] [Accepted: 05/03/2019] [Indexed: 02/05/2023]
Abstract
Molecular imaging techniques apply sophisticated technologies to monitor, directly or indirectly, the spatiotemporal distribution of molecular or cellular processes for biomedical, diagnostic, or therapeutic purposes. For example, Single-Photon Emission Computed Tomography (SPECT) and Positron Emission Tomography (PET) imaging, the most representative modalities of molecular imaging, enable earlier and more accurate diagnosis of cancer and cardiovascular diseases. New possibilities for noninvasive molecular imaging in vivo have emerged with advances in bioorthogonal chemistry. For example, tetrazine-related Inverse Electron Demand Diels-Alder (IEDDA) reactions can rapidly generate short-lived radioisotope probes in vivo that provide strong contrast for SPECT and PET. Here, we review pretargeting strategies for molecular imaging and novel radiotracers synthesized via tetrazine bioorthogonal chemistry. We systematically describe advances in direct radiolabeling and pretargeting approaches in SPECT and PET using metal and nonmetal radioisotopes based on tetrazine bioorthogonal reactions, and we discuss prospects for the future of such contrast agents.
Collapse
Affiliation(s)
- Ping Dong
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xueyi Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Junwei Zheng
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaoyang Zhang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yiwen Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Haoxing Wu
- Huaxi MR Research Center, Department of Radiology, West China Hospital and West China School of Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lin Li
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
50
|
Zettlitz KA, Waldmann CM, Tsai WTK, Tavaré R, Collins J, Murphy JM, Wu AM. A Dual-Modality Linker Enables Site-Specific Conjugation of Antibody Fragments for 18F-Immuno-PET and Fluorescence Imaging. J Nucl Med 2019; 60:1467-1473. [PMID: 30877181 DOI: 10.2967/jnumed.118.223560] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/06/2019] [Indexed: 12/30/2022] Open
Abstract
Antibody-based dual-modality (PET/fluorescence) imaging enables both presurgery antigen-specific immuno-PET for noninvasive whole-body evaluation and intraoperative fluorescence for visualization of superficial tissue layers for image-guided surgery. Methods: We developed a universal dual-modality linker (DML) that facilitates site-specific conjugation to a cysteine residue-bearing antibody fragment, introduction of a commercially available fluorescent dye (via an amine-reactive prosthetic group), and rapid and efficient radiolabeling via click chemistry with 18F-labeled trans-cyclooctene (18F-TCO). To generate a dual-modality antibody fragment-based imaging agent, the DML was labeled with the far-red dye sulfonate cyanine 5 (sCy5), site-specifically conjugated to the C-terminal cysteine of the anti-prostate stem cell antigen (PSCA) cys-diabody A2, and subsequently radiolabeled by click chemistry with 18F-TCO. The new imaging probe was evaluated in a human PSCA-positive prostate cancer xenograft model by sequential immuno-PET and optical imaging. Uptake in target tissues was confirmed by ex vivo biodistribution. Results: We successfully synthesized a DML for conjugation of a fluorescent dye and 18F. The anti-PSCA cys-diabody A2 was site-specifically conjugated with either DML or sCy5 and radiolabeled via click chemistry with 18F-TCO. Immuno-PET imaging confirmed in vivo antigen-specific targeting of prostate cancer xenografts as early as 1 h after injection. Rapid renal clearance of the 50-kDa antibody fragment enables same-day imaging. Optical imaging showed antigen-specific fluorescent signal in PSCA-positive xenografts and high contrast to surrounding tissue and PSCA-negative xenografts. Conclusion: The DML enables site-specific conjugation away from the antigen-binding site of antibody fragments, with a controlled linker-to-protein ratio, and combines signaling moieties for 2 imaging systems into 1 molecule. Dual-modality imaging could provide both noninvasive whole-body imaging with organ-level biodistribution and fluorescence image-guided identification of tumor margins during surgery.
Collapse
Affiliation(s)
- Kirstin A Zettlitz
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Christopher M Waldmann
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Wen-Ting K Tsai
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Richard Tavaré
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Jeffrey Collins
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Jennifer M Murphy
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Anna M Wu
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|