1
|
Gamelas SRD, Shmuel S, Simó C, Vanover A, Tomé JPC, Tomé AC, Lourenço LMO, Pereira PMR. Positron Emission Tomography and Optical Imaging to Monitor Bioorthogonal Diels-Alder Click Chemistry of Trastuzumab with a Porphyrin. Bioconjug Chem 2025; 36:1013-1020. [PMID: 40353799 DOI: 10.1021/acs.bioconjchem.5c00056] [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: 05/14/2025]
Abstract
Click chemistry to allow in vivo conjugation of a fluorophore porphyrin (Por)-tetrazine (Tz) with the human epidermal growth factor receptor 2 (HER2)-targeting trastuzumab conjugated with trans-cyclooctene (TCO) is described here. In vitro experiments confirmed successful click reactions between Por-Tz and trastuzumab-TCO and validated preserved trastuzumab immunoreactivity (no significant change in HER2 binding, p > 0.05). Positron emission tomography (PET) of [89Zr]Zr-DFO-trastuzumab-TCO demonstrated 17.1 ± 2.9% injected dose per gram of tumor at 48 h postinjection. Optical imaging showed an ∼10-fold increase in the click group for Por-Tz when compared with Por-Tz alone. This preclinical data demonstrate a pretargeted approach for dual PET and optical imaging of HER2-expressing tumors.
Collapse
Affiliation(s)
- Sara R D Gamelas
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
- LAQV-REQUIMTE, Departamento de Química, Universidade de Aveiro, Aveiro 3810-193, Portugal
| | - Shayla Shmuel
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Cristina Simó
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Alex Vanover
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - João P C Tomé
- CQE, Institute of Molecular Sciences, Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal
| | - Augusto C Tomé
- LAQV-REQUIMTE, Departamento de Química, Universidade de Aveiro, Aveiro 3810-193, Portugal
| | - Leandro M O Lourenço
- LAQV-REQUIMTE, Departamento de Química, Universidade de Aveiro, Aveiro 3810-193, Portugal
| | - Patrícia M R Pereira
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| |
Collapse
|
2
|
Hvass L, Müller M, Staudt M, García-Vázquez R, Gustavsson TK, Shalgunov V, Jørgensen JT, Battisti UM, Herth MM, Kjaer A. Head-to-Head Comparison of the in Vivo Performance of Highly Reactive and Polar 18F-Labeled Tetrazines. Mol Pharm 2025; 22:1911-1919. [PMID: 40081392 PMCID: PMC11979891 DOI: 10.1021/acs.molpharmaceut.4c01129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 03/16/2025]
Abstract
Pretargeted imaging harnessing tetrazine ligation has gained increased interest over recent years. Targeting vectors with slow pharmacokinetics may be visualized using short-lived radionuclides, such as fluorine-18 (18F) for positron emission tomography (PET), and result in improved target-to-background ratios compared to conventionally radiolabeled slowly accumulating vectors. We recently developed different radiochemical protocols enabling the direct radiofluorination of various tetrazine scaffolds, resulting in the development of various highly reactive and polar 18F-labeled tetrazines as lead candidates for pretargeted imaging. Here, we performed a direct head-to-head-comparison of our lead candidates to evaluate the most promising for future clinical translation. For that, all 18F-labeled tetrazine-scaffolds were synthesized in similar molar activity for improved comparability of their in vivo pretargeting performance. Intriguingly, previously reported dicarboxylic acid lead candidates with a net charge of -1 were outperformed by respective monocarboxylic acid derivatives bearing a net charge of 0, warranting further evaluation of such scaffolds prior to their clinical translation.
Collapse
Affiliation(s)
- Lars Hvass
- Cluster
for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen N, Denmark
- Department
of Clinical Physiology, Nuclear Medicine
& PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Marius Müller
- Department
of Clinical Physiology, Nuclear Medicine
& PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Markus Staudt
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Rocio García-Vázquez
- Department
of Clinical Physiology, Nuclear Medicine
& PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Tobias K. Gustavsson
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Vladimir Shalgunov
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Jesper T. Jørgensen
- Cluster
for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen N, 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, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Matthias M. Herth
- Department
of Clinical Physiology, Nuclear Medicine
& PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Andreas Kjaer
- Cluster
for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen N, Denmark
- Department
of Clinical Physiology, Nuclear Medicine
& PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| |
Collapse
|
3
|
Sarkar S, Pham JM, Edwards KJ, Sharma N, Xu K, King AP, Del Castillo AF, Farwell MD, Pryma DA, Schuster SJ, Sellmyer MA. A biorthogonal chemistry approach for high-contrast antibody imaging of lymphoma at early time points. EJNMMI Res 2025; 15:26. [PMID: 40122966 PMCID: PMC11930911 DOI: 10.1186/s13550-025-01213-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: 01/04/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Monoclonal antibodies are highly specific for their targets making them effective for cancer therapy. However, their large molecular weight causes slow blood clearance, often requiring weeks to be removed from circulation. This limitation affects companion nuclear imaging and antibody-based diagnostics, necessitating delayed imaging. We report the expansion of a methodology improving positron emission tomography (PET) contrast of the lymphoma biomarker CD20 at early time points after radiolabeled antibody administration. Intact radioimmunoconjugates are allowed to stay in circulation long enough to accumulate in tumors, and then, using a chemical trigger, we induced rapid clearance of the radioactivity from non-target tissues by cleaving the linker between the antibody and the radioactivity. For brevity, we refer to the this as the Tetrazine KnockOut (TKO) method which uses the transcyclooctene-tetrazine (TCO-Tz) reaction, wherein an antibody is conjugated with linker containing TCO and a radioisotope. RESULTS We optimized the TCO linker with several different radioisotopes and evaluated the ability of tetrazines to knockout radioactivity from circulating antibodies. We explored several cell types and antibodies with varying internalization rates, to characterize the parameters of TKO and tested [89Zr]Zr-DFO-TCO-rituximab in a lymphoma model with PET imaging after Tz or vehicle administration. Treatment with Tz induced > 70% cleavage of the TCO linker in vitro within 30 min. Internalizing radioimmunoconjugates exhibited similar cellular uptake with Tz compared to vehicle, whereas decreased uptake was seen with slowly internalizing antibodies. In rodents, Tz rapidly liberated the radioactivity from the antibody, cleared from the blood, and accumulated in the bladder. TKO resulted in > 50% decreased radioactivity in non-target organs following Tz injection. No decrease in tumor uptake was observed when rate of antibody internalization is higher in a lymphoma model, and the target-to-background ratio increased by > twofold in comparison with Tz nontreated groups at 24 h. CONCLUSION The TKO approach potentiates early imaging of rituximab radioimmunoconjugates and has translational potential for lymphoma imaging.
Collapse
Affiliation(s)
- Swarbhanu Sarkar
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA
| | - Jonathan M Pham
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA
| | - Kimberly J Edwards
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA
| | - Nitika Sharma
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA
| | - Kexiang Xu
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA
| | - A Paden King
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA
| | - Andres Fernandez Del Castillo
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA
| | - Michael D Farwell
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA
| | - Daniel A Pryma
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA
| | - Stephen J Schuster
- Lymphoma Program, Abramson Cancer Center at the University of Pennsylvania, Philadelphia, PA, USA
| | - Mark A Sellmyer
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 813A Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA, 19104-6059, USA.
- The Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
4
|
de Roode KE, Rossin R, Robillard MS. Toward Realization of Bioorthogonal Chemistry in the Clinic. Top Curr Chem (Cham) 2025; 383:12. [PMID: 40042792 PMCID: PMC11882664 DOI: 10.1007/s41061-025-00495-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025]
Abstract
In the last decade, the use of bioorthogonal chemistry toward medical applications has increased tremendously. Besides being useful for the production of pharmaceuticals, the efficient, nontoxic reactions open possibilities for the development of therapies that rely on in vivo chemistry between two bioorthogonal components. Here we discuss the latest developments in bioorthogonal chemistry, with a focus on their use in living organisms, the translation from model systems to humans, and the challenges encountered during preclinical development. We aim to provide the reader a broad presentation of the current state of the art and demonstrate the numerous possibilities that bioorthogonal reactions have for clinical use, now and in the near future.
Collapse
Affiliation(s)
- Kim E de Roode
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED, Nijmegen, The Netherlands
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Raffaella Rossin
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED, Nijmegen, The Netherlands
| | - Marc S Robillard
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED, Nijmegen, The Netherlands.
| |
Collapse
|
5
|
Herth MM, Hvass L, Poulie CBM, Müller M, García-Vázquez R, Gustavsson T, Shalgunov V, Clausen AS, Jørgensen JT, Hansson E, Jensen H, Aneheim E, Lindegren S, Kjaer A, Battisti UM. An 211At-labeled Tetrazine for Pretargeted Therapy. J Med Chem 2025; 68:4410-4425. [PMID: 39963966 DOI: 10.1021/acs.jmedchem.4c02281] [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
Pretargeted radioimmunoimaging has been shown to enhance tumor-to-background ratios by up to 125-fold at early time points, leading to more efficient and less toxic radionuclide therapies, particularly with shorter half-lives such as astatine-211 (211At). The tetrazine ligation is the most utilized bioorthogonal reaction in these strategies, making tetrazines ideal for 211At labeling and controlling the biodistribution. We developed a 211At-labeled pretargeting agent for alpha-radionuclide therapy, achieving a radiochemical yield of approximately 65% and purity over 99%. Our results showed higher tumor-to-blood ratios within the first 24 h compared to directly labeled monoclonal antibodies. This suggests that pretargeted therapy may deliver better tumor doses than conventional methods, although the deastatination observed will need to be addressed in future tetrazine developments.
Collapse
Affiliation(s)
- Matthias M Herth
- 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, Cyclotron and Radiochemistry Unit, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Christian B M Poulie
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Marius Müller
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Rocio García-Vázquez
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Tobias Gustavsson
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Vladimir Shalgunov
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen Ø, Denmark
| | - Anne S Clausen
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen Ø, Denmark
- Department of Clinical Physiology, Nuclear Medicine, Cyclotron and Radiochemistry Unit, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Jesper T Jørgensen
- Department of Clinical Physiology, Nuclear Medicine, Cyclotron and Radiochemistry Unit, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Ellinor Hansson
- Departments of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gula Stråket 2b, 41345 Gothenburg, Sweden
| | - Holger Jensen
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen Ø, Denmark
| | - Emma Aneheim
- Departments of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gula Stråket 2b, 41345 Gothenburg, Sweden
| | - Sture Lindegren
- Departments of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gula Stråket 2b, 41345 Gothenburg, Sweden
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine, Cyclotron and Radiochemistry Unit, Rigshospitalet, 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
|
6
|
Li J, Cheng M, Zhang H, Wang Y, Guo W, Zheng Y. A Tetrazine Amplification System for Visual Detection of Trace Analytes via Click-Release Reactions. Angew Chem Int Ed Engl 2025; 64:e202414246. [PMID: 39623886 DOI: 10.1002/anie.202414246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 12/02/2024] [Indexed: 12/17/2024]
Abstract
Achieving visual detection of analytes at ultra-low concentrations in complex mixtures remains a persistent challenge. While sophisticated techniques offer single-molecule sensitivity, practical hurdles remain, necessitating tailored signal amplification systems for direct visual detection. In this study, we develop a strategy for the visualized detection of tetrazine through a "click-release-oxidation-cycle" (CROC) cascade amplification process. We systematically describe the construction and synthesis of this system, the kinetic process of click release, the kinetics of oxidation to tetrazine and its cascade amplification effect in trace amounts of tetrazine. This system is capable of amplifying the signal of tetrazine at a concentration as low as 2 nM by 105-fold, thereby providing a clearly visible purple signal. Finally, as proof of concept, we successfully apply this method to visually detect trace β-galactosidase (β-gal) and Pd2+.
Collapse
Affiliation(s)
- Jing Li
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Mingxin Cheng
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Hongbo Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Yichen Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Weiwei Guo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 211198, P. R. China
- School of Science, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Yueqin Zheng
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 211198, P. R. China
| |
Collapse
|
7
|
Hai W, Bao X, Sun K, Li B, Peng J, Xu Y. In situ labeling of pretargeted hyaluronan for PET/MR imaging of CD44+ tumors. Bioorg Chem 2025; 155:108110. [PMID: 39756203 DOI: 10.1016/j.bioorg.2024.108110] [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: 09/14/2024] [Revised: 12/13/2024] [Accepted: 12/28/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Tumor-specific molecular probe-based imaging strategies have shown great potential for tumor diagnosis. However, the sensitivity and contrast of imaging may interfere with the complex labeling process and degradation of tumor-specific imaging probes. We sought to adapt a pretargeting strategy and an in vivo bioorthogonal reaction to improve hyaluronan (HA)-based tumor multimodal imaging diagnosis. METHODS Transcyclooctene-labeled HA (HA-TCO) and tetrazine-labeled NODA (NODA-Tz) were synthesized and purified. Probes Gd-NODA-Tz and [18F]AlF-NODA-Tz for magnetic resonance imaging (MRI) and positron emission tomography (PET) imaging were prepared. The bioorthogonal reaction of HA-TCO with NODA-Tz and the stability of the products were confirmed and analyzed. CD44 + A549 tumor-bearing mice were injected with HA-TCO via the tail vein, followed by Gd-NODA-Tz or [18F]AlF-NODA-Tz administration half an hour later, and subsequently imaged by MR or PET. The images were analyzed and tumor uptake was quantified. RESULTS HA-TCO efficiently bound to CD44-overexpressing A549 cells and selectively reacted with the Tz-imaging group. In vivo MR and PET images were obtained after probe injection and subsequent bioorthogonal labeling. The images showed a tumor mass with a high target background ratio (TBR) and clear boundaries. CONCLUSION In situ labeling of pretargeted HA-TCO enabled MRI and PET imaging of tumor tissues in mice with high sensitivity and improved TBR.
Collapse
Affiliation(s)
- Wangxi Hai
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China; School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Xiao Bao
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Kang Sun
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China.
| | - Jinliang Peng
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Yuhong Xu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China; College of Pharmacy, Dali University, Xia Guan, Dali, Yunnan 6710000, PR China.
| |
Collapse
|
8
|
Mack KN, Bauer D, Carter LM, Carrasco SE, Atmane MI, Viray TD, Brooks CL, Hollingsworth MA, Radhakrishnan P, Lewis JS. Pretargeted alpha therapy in MUC16-positive high-grade serous ovarian cancer. Nucl Med Biol 2025; 140-141:108976. [PMID: 39615062 DOI: 10.1016/j.nucmedbio.2024.108976] [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: 09/06/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 03/15/2025]
Abstract
BACKGROUND Peritoneal metastasis with micrometastatic cell clusters is a common feature of advanced ovarian cancer. Targeted alpha therapy (TAT) is an attractive approach for treating micrometastatic diseases as alpha particles release enormous amounts of energy within a short distance. A pretargeting approach - leveraging the inverse-electron-demand Diels-Alder reaction between tetrazines (Tz) and trans-cyclooctene (TCO) - can minimize off-target toxicity related to TAT, often associated with full-length antibodies. We hypothesized that a pretargeting strategy could effectively treat high-grade serous (HGS) ovarian tumors while minimizing toxicity. METHODS We utilized the humanized antibody, AR9.6, labeled with actinium-225 (225Ac). AR9.6 targets fully glycosylated and hypoglycosylated isoforms of MUC16. For biodistribution and radioimmunotherapy studies, AR9.6-TCO was injected into OVCAR3-bearing mice 72 h before administering [225Ac]Ac-mcp-PEG8-Tz, e.g. using a 1,2,4,5-tetrazine conjugated to the macropa chelator via a polyethylene glycol (PEG) linker. RESULTS Biodistribution data revealed that the pretargeting approach achieved substantial tumor uptake. Cerenkov luminescence imaging confirmed successful in vivo pretargeting during TAT studies. Compared to the control groups, TAT with AR9.6-TCO and [225Ac]Ac-mcp-PEG8-Tz significantly suppressed tumor growth and improved overall survival in OVCAR3 tumor-bearing mice. Renal and ovarian pathology compatible with toxicity was observed in mice in addition to transient hematologic toxicity. CONCLUSION We confirmed that pretargeting with AR9.6-TCO and [225Ac]Ac-mcp-PEG8-Tz has durable antitumor effects in high MUC16-expressing tumors. These findings demonstrate great potential for using pretargeting in combination with TAT for the treatment of ovarian cancer. CLASSIFICATION Biological Sciences; Applied Biological Sciences.
Collapse
Affiliation(s)
- Kyeara N Mack
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Pharmacology, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - David Bauer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lukas M Carter
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sebastian E Carrasco
- Laboratory of Comparative Pathology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, and The Rockefeller University, NY, New York, USA
| | - Mohamed I Atmane
- Laboratory of Comparative Pathology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, and The Rockefeller University, NY, New York, USA
| | - Tara D Viray
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cory L Brooks
- Department of Chemistry and Biochemistry, California State University, Fresno, CA, USA
| | - Michael A Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, NE, USA
| | - Prakash Radhakrishnan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, NE, USA
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Pharmacology, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA; Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
9
|
Abbas SJ, Yesmin S, Vittala SK, Sepay N, Xia F, Ali SI, Chang WC, Hung YC, Ma WL. Target Bioconjugation of Protein Through Chemical, Molecular Dynamics, and Artificial Intelligence Approaches. Metabolites 2024; 14:668. [PMID: 39728449 DOI: 10.3390/metabo14120668] [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: 10/02/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
Covalent modification of proteins at specific, predetermined sites is essential for advancing biological and biopharmaceutical applications. Site-selective labeling techniques for protein modification allow us to effectively track biological function, intracellular dynamics, and localization. Despite numerous reports on modifying target proteins with functional chemical probes, unique organic reactions that achieve site-selective integration without compromising native functional properties remain a significant challenge. In this review, we delve into site-selective protein modification using synthetic probes, highlighting both chemical and computational methodologies for chemo- and regioselective modifications of naturally occurring amino acids, as well as proximity-driven protein-selective chemical modifications. We also underline recent traceless affinity labeling strategies that involve exchange/cleavage reactions and catalyst tethering modifications. The rapid development of computational infrastructure and methods has made the bioconjugation of proteins more accessible, enabling precise predictions of structural changes due to protein modifications. Hence, we discuss bioconjugational computational approaches, including molecular dynamics and artificial intelligence, underscoring their potential applications in enhancing our understanding of cellular biology and addressing current challenges in the field.
Collapse
Affiliation(s)
- Sk Jahir Abbas
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Department of Obstetrics and Gynecology, Asia University Hospital, Taichung 41354, Taiwan
| | - Sabina Yesmin
- Institute of Chemistry, Academia Sinica, Taipei 115201, Taiwan
- Department of Physics, National Dong Hwa University, Hualien 97401, Taiwan
| | - Sandeepa K Vittala
- Leiden Institute of Chemistry, Leiden University, 2300 RA Leiden, The Netherlands
| | - Nayim Sepay
- Department of Chemistry, Lady Brabourne College, Kolkata 700017, India
| | - Fangfang Xia
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Centre, Houston, TX 77030, USA
| | - Sk Imran Ali
- Department of Chemistry, University of Kalyani, Kalyani 741235, India
| | - Wei-Chun Chang
- Ph.D. Program for Health Science and Industry, China Medical University, Taichung 40402, Taiwan
- Department of Medical Research, Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung 40402, Taiwan
| | - Yao-Ching Hung
- Department of Obstetrics and Gynecology, Asia University Hospital, Taichung 41354, Taiwan
| | - Wen-Lung Ma
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Department of Medical Research, Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung 40402, Taiwan
| |
Collapse
|
10
|
Poty S, Ordas L, Dekempeneer Y, Parach AA, Navarro L, Santens F, Dumauthioz N, Bardiès M, Lahoutte T, D'Huyvetter M, Pouget JP. Optimizing the Therapeutic Index of sdAb-Based Radiopharmaceuticals Using Pretargeting. J Nucl Med 2024; 65:1564-1570. [PMID: 39266288 PMCID: PMC11448608 DOI: 10.2967/jnumed.124.267624] [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: 02/16/2024] [Accepted: 06/05/2024] [Indexed: 09/14/2024] Open
Abstract
Single-domain antibodies (sdAbs) demonstrate favorable pharmacokinetic profiles for molecular imaging applications. However, their renal excretion and retention are obstacles for applications in targeted radionuclide therapy (TRT). Methods: Using a click-chemistry-based pretargeting approach, we aimed to reduce kidney retention of a fibroblast activation protein α (FAP)-targeted sdAb, 4AH29, for 177Lu-TRT. Key pretargeting parameters (sdAb-injected mass and lag time) were optimized in healthy mice and U87MG (FAP+) xenografts. A TRT study in a pancreatic ductal adenocarcinoma (PDAC) patient-derived xenograft (PDX) model was performed as a pilot study for sdAb-based pretargeting applications. Results: Modification of 4AH29 with trans-cyclooctene (TCO) moieties did not modify the sdAb pharmacokinetic profile. A 200-µg injected mass of 4AH29-TCO and an 8-h lag time for the injection of [177Lu]Lu-DOTA-PEG7-tetrazine resulted in the highest kidney therapeutic index (2.0 ± 0.4), which was 5-fold higher than that of [177Lu]Lu-DOTA-4AH29 (0.4 ± 0.1). FAP expression in the tumor microenvironment was validated in a PDAC PDX model with both immunohistochemistry and PET/CT imaging. Mice treated with the pretargeting high-activity approach (4AH29-TCO + [177Lu]Lu-DOTA-PEG7-tetrazine; 3 × 88 MBq, 1 injection per week for 3 wk) demonstrated prolonged survival compared with the vehicle control and conventionally treated ([177Lu]Lu-DOTA-4AH29; 3 × 37 MBq, 1 injection per week for 3 wk) mice. Mesangial expansion was reported in 7 of 10 mice in the conventional cohort, suggesting treatment-related kidney morphologic changes, but was not observed in the pretargeting cohort. Conclusion: This study validates pretargeting to mitigate sdAbs' kidney retention with no observation of morphologic changes on therapy regimen at early time points. Clinical translation of click-chemistry-based pre-TRT is warranted on the basis of its ability to alleviate toxicities related to biovectors' intrinsic pharmacokinetic profiles. The absence of representative animal models with extensive stroma and high FAP expression on cancer-associated fibroblasts led to a low mean tumor-absorbed dose even with high injected activity and consequently to modest survival benefit in this PDAC PDX.
Collapse
Affiliation(s)
- Sophie Poty
- Institut de Recherche en Cancérologie de Montpellier, Université de Montpellier, INSERM, U1194, Équipe labellisée Ligue contre le cancer, Montpellier, France; and
| | - Laura Ordas
- Institut de Recherche en Cancérologie de Montpellier, Université de Montpellier, INSERM, U1194, Équipe labellisée Ligue contre le cancer, Montpellier, France; and
| | | | - Ali Asghar Parach
- Institut de Recherche en Cancérologie de Montpellier, Université de Montpellier, INSERM, U1194, Équipe labellisée Ligue contre le cancer, Montpellier, France; and
| | | | | | | | - Manuel Bardiès
- Institut de Recherche en Cancérologie de Montpellier, Université de Montpellier, INSERM, U1194, Équipe labellisée Ligue contre le cancer, Montpellier, France; and
| | | | | | - Jean-Pierre Pouget
- Institut de Recherche en Cancérologie de Montpellier, Université de Montpellier, INSERM, U1194, Équipe labellisée Ligue contre le cancer, Montpellier, France; and
| |
Collapse
|
11
|
Huang Y, Li Z, Li C, Zang Z, Wang Q, Huang S, Liu Q, Liang Y. Bioorthogonal Diels-Alder Click Chemistry-Based Pretargeted PET Imaging Strategy for Monitoring Programmed Death-Ligand 1 Expression. ACS OMEGA 2024; 9:36969-36981. [PMID: 39246495 PMCID: PMC11375721 DOI: 10.1021/acsomega.4c01063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 07/05/2024] [Accepted: 08/14/2024] [Indexed: 09/10/2024]
Abstract
The development of antibody tracers for positron emission tomography (PET) imaging enables real-time monitoring of tumor expression of programmed cell death ligand 1 (PD-L1) in vivo, aiming to facilitate the selection of immunotherapy responders. However, the slow pharmacokinetics of the antibodies in vivo limits their applications in PET imaging with commonly used radionuclides with short half-lives. In this study, we developed a pretargeted PET imaging strategy based on Diels-Alder (IEDDA) click chemistry to overcome these limitations. Atezolizumab and durvalumab, the most commonly used PD-L1 antibodies in cancer immunotherapy, were selected and compared in the development of the pretargeted PET imaging strategy. Fluorine-18-labeled derivatives of methyl tetrazine ([18F]Tz, [18F]PEG6-Tz, and [18F]PEG12-Tz) were tested in biodistribution and PET imaging of A549-PDL1 xenografts (PD-L1 positive) pretargeted with the trans-cyclooctene (TCO)-functionalized atezolizumab/durvalumab. The biodistribution and imaging results indicated that atezolizumab-TCO/[18F]PEG12-Tz was more suitable for pretargeted PET imaging strategy, and the optimal interval time was 48 h after atezolizumab-TCO administration, where the atezolizumab-TCO/[18F]PEG12-Tz pretargeted approach clearly delineated the A549-PDL1 tumor with a tumor-to-muscle ratio of 5.33, while the ratios are 3.39 and 2.39 for durvalumab/[18F]PEG12-Tz and mock-pretargeting controls, respectively. In conclusion, a pretargeted 18F-immuno-PET imaging technology was successfully established on atezolizumab. The high-contrast PET images of the A549-PDL1 tumor models demonstrate that the pretargeting strategy incorporating short-lived fluorine-18 is viable in identifying tumors with high PD-L1 expression, marking this strategy as a potential candidate for further clinical translation.
Collapse
Affiliation(s)
- Yong Huang
- Department of Nuclear Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Zhongjing Li
- Department of Nuclear Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Chengze Li
- Department of Nuclear Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Zihan Zang
- Shenzhen Middle School, Shenzhen 518024, China
| | - Qiong Wang
- Department of Nuclear Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Size Huang
- Department of Nuclear Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Qi Liu
- International Cancer Center, Shenzhen University School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Ying Liang
- Department of Nuclear Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| |
Collapse
|
12
|
Mishra A, Carrascal-Miniño A, Kim J, T M de Rosales R. [ 68Ga]Ga-THP-tetrazine for bioorthogonal click radiolabelling: pretargeted PET imaging of liposomal nanomedicines. RSC Chem Biol 2024; 5:622-639. [PMID: 38966673 PMCID: PMC11221536 DOI: 10.1039/d4cb00039k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/10/2024] [Indexed: 07/06/2024] Open
Abstract
Pretargeted PET imaging using bioorthogonal chemistry is a leading strategy for the tracking of long-circulating agents such as antibodies and nanoparticle-drug delivery systems with short-lived isotopes. Here, we report the synthesis, characterisation and in vitro/vivo evaluation of a new 68Ga-based radiotracer [68Ga]Ga-THP-Tetrazine ([68Ga]Ga-THP-Tz) for bioorthogonal click radiochemistry and in vivo labelling of agents with slow pharmacokinetics. THP-tetrazine (THP-Tz) can be radiolabelled to give [68/67Ga]Ga-THP-Tz at room temperature in less than 15 minutes with excellent radiochemical stability in vitro and in vivo. [68Ga]Ga-THP-Tz was tested in vitro and in vivo for pretargeted imaging of stealth PEGylated liposomes, chosen as a leading clinically-approved platform of nanoparticle-based drug delivery, and for their known long-circulating properties. To achieve this, PEGylated liposomes were functionalised with a synthesised transcyclooctene (TCO) modified phospholipid. Radiolabelling of TCO-PEG-liposomes with [68/67Ga]Ga-THP-Tz was demonstrated in vitro in human serum, and in vivo using both healthy mice and in a syngeneic cancer murine model (WEHI-164 fibrosarcoma). Interestingly in vivo data revealed that [68Ga]Ga-THP-Tz was able to in vivo radiolabel liposomes present in the liver and spleen, and not those in the blood pool or in the tumour. Overall, these results demonstrate the potential of [68Ga]Ga-THP-Tz for pretargeted imaging/therapy but also some unexpected limitations of this system.
Collapse
Affiliation(s)
- Aishwarya Mishra
- School of Biomedical Engineering & Imaging Sciences, King's College London St Thomas' Hospital London SE1 7EH UK
| | - Amaia Carrascal-Miniño
- School of Biomedical Engineering & Imaging Sciences, King's College London St Thomas' Hospital London SE1 7EH UK
| | - Jana Kim
- School of Biomedical Engineering & Imaging Sciences, King's College London St Thomas' Hospital London SE1 7EH UK
| | - Rafael T M de Rosales
- School of Biomedical Engineering & Imaging Sciences, King's College London St Thomas' Hospital London SE1 7EH UK
| |
Collapse
|
13
|
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
|
14
|
Alanizi AA, Sorlin AM, Parker MFL, López-Álvarez M, Qin H, Lee SH, Blecha J, Rosenberg OS, Engel J, Ohliger MA, Flavell RR, Wilson DM. Bioorthogonal Radiolabeling of Azide-Modified Bacteria Using [ 18F]FB-sulfo-DBCO. Bioconjug Chem 2024; 35:517-527. [PMID: 38482815 PMCID: PMC11036355 DOI: 10.1021/acs.bioconjchem.4c00024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 04/18/2024]
Abstract
Purpose: This study was motivated by the need for better positron emission tomography (PET)-compatible tools to image bacterial infection. Our previous efforts have targeted bacteria-specific metabolism via assimilation of carbon-11 labeled d-amino acids into the bacterial cell wall. Since the chemical determinants of this incorporation are not fully understood, we sought a high-throughput method to label d-amino acid derived structures with fluorine-18. Our strategy employed a chemical biology approach, whereby an azide (-N3) bearing d-amino acid is incorporated into peptidoglycan muropeptides, with subsequent "click" cycloaddition with an 18F-labeled strained cyclooctyne partner. Procedures: A water-soluble, 18F-labeled and dibenzocyclooctyne (DBCO)-derived radiotracer ([18F]FB-sulfo-DBCO) was synthesized. This tracer was incubated with pathogenic bacteria treated with azide-bearing d-amino acids, and incorporated 18F was determined via gamma counting. In vitro uptake in bacteria previously treated with azide-modified d-amino acids was compared to that in cultures treated with amino acid controls. The biodistribution of [18F]FB-sulfo-DBCO was studied in a cohort of healthy mice with implications for future in vivo imaging. Results: The new strain-promoted azide-alkyne cycloaddition (SPAAC) radiotracer [18F]FB-sulfo-DBCO was synthesized with high radiochemical yield and purity via N-succinimidyl 4-[18F]fluorobenzoate ([18F]SFB). Accumulation of [18F]FB-sulfo-DBCO was significantly higher in several bacteria treated with azide-modified d-amino acids than in controls; for example, we observed 7 times greater [18F]FB-sulfo-DBCO ligation in Staphylococcus aureus cultures incubated with 3-azido-d-alanine versus those incubated with d-alanine. Conclusions: The SPAAC radiotracer [18F]FB-sulfo-DBCO was validated in vitro via metabolic labeling of azide-bearing peptidoglycan muropeptides. d-Amino acid-derived PET radiotracers may be more efficiently screened via [18F]FB-sulfo-DBCO modification.
Collapse
Affiliation(s)
- Aryn A. Alanizi
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
| | - Alexandre M. Sorlin
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
| | - Matthew F. L. Parker
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
- Department
of Psychiatry, Renaissance School of Medicine
at Stony Brook University, Stony
Brook, New York 11794, United States
| | - Marina López-Álvarez
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
| | - Hecong Qin
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
| | - Sang Hee Lee
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
| | - Joseph Blecha
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
| | - Oren S. Rosenberg
- Department
of Medicine, University of California, San
Francisco, San Francisco, California 94158, United States
| | - Joanne Engel
- Department
of Medicine, University of California, San
Francisco, San Francisco, California 94158, United States
| | - Michael A. Ohliger
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
- Department
of Radiology, Zuckerberg San Francisco General
Hospital, San Francisco, California 94110, United States
| | - Robert R. Flavell
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
| | - David M. Wilson
- Department
of Radiology and Biomedical Imaging, University
of California, San Francisco, San Francisco, California 94158, United States
| |
Collapse
|
15
|
Bohrmann L, Poulie CBM, Rodríguez-Rodríguez C, Karagiozov S, Saatchi K, Herth MM, Häfeli UO. Development of a 99mTc-labeled tetrazine for pretargeted SPECT imaging using an alendronic acid-based bone targeting model. PLoS One 2024; 19:e0300466. [PMID: 38626058 PMCID: PMC11020896 DOI: 10.1371/journal.pone.0300466] [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: 08/10/2023] [Accepted: 02/27/2024] [Indexed: 04/18/2024] Open
Abstract
Pretargeting, which is the separation of target accumulation and the administration of a secondary imaging agent into two sequential steps, offers the potential to improve image contrast and reduce radiation burden for nuclear imaging. In recent years, the tetrazine ligation has emerged as a promising approach to facilitate covalent pretargeted imaging due to its unprecedented kinetics and bioorthogonality. Pretargeted bone imaging with TCO-modified alendronic acid (Aln-TCO) is an attractive model that allows the evaluation of tetrazines in healthy animals without the need for complex disease models or targeting regimens. Recent structure-activity relationship studies of tetrazines evaluated important parameters for the design of potent tetrazine-radiotracers for pretargeted imaging. However, limited information is available for 99mTc-labeled tetrazines. In this study, four tetrazines intended for labeling with fac-[99mTc(OH2)3 (CO)3]+ were synthesized and evaluated using an Aln-TCO mouse model. 3,6-bis(2-pyridyl)-1,2,4,5-Tz without additional linker showed higher pretargeted bone uptake and less background activity compared to the same scaffold with a PEG8 linker or 3-phenyl-1,2,4,5-Tz-based compounds. Additionally, improved bone/blood ratios were observed in pretargeted animals compared to animals receiving directly labeled Aln-TCO. The results of this study implicate 3,6-bis(2-pyridyl)-1,2,4,5-Tz as a promising scaffold for potential 99mTc-labeled tetrazines.
Collapse
Affiliation(s)
- Lennart Bohrmann
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken, Copenhagen, Denmark
| | - Christian B. M. Poulie
- Department of Drug Design and Pharmacology, Faculty of Health and Medicinal Sciences, University of Copenhagen, Universitetsparken, Copenhagen, Denmark
| | | | - Stoyan Karagiozov
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Katayoun Saatchi
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Matthias M. Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medicinal Sciences, University of Copenhagen, Universitetsparken, Copenhagen, Denmark
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej, Copenhagen, Denmark
| | - Urs O. Häfeli
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken, Copenhagen, Denmark
| |
Collapse
|
16
|
Zentel R. Nanoparticular Carriers As Objects to Study Intentional and Unintentional Bioconjugation. ACS Biomater Sci Eng 2024; 10:3-11. [PMID: 35412796 DOI: 10.1021/acsbiomaterials.2c00091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Synthetic nanoparticles are interesting to use in the study of ligation with natural biorelevant structures. That is because they present an intermediate situation between reactions onto soluble polymers or onto solid surfaces. In addition, differently functionalized nanoparticles can be separated and studied independently thereafter. So what would be a "patchy functionalization" on a macroscopic surface results in differently functionalized nanoparticles, which can be separated after the interaction with body fluids. This paper will review bioconjugation of such nanoparticles with a special focus on recent results concerning the formation of a protein corona by unspecific adsorption (lower lines of TOC), which presents an unintentional bioconjugation, and on new aspects of intentionally performed bioconjugation by covalent chemistry (upper line). For this purpose, it is important that polymeric nanoparticles without a protein corona can be prepared. This opens, e.g., the possibility to look for special proteins adsorbed as a result of the natural compound ligated to the nanoparticle by covalent chemistry, like the Fc part of antibodies. At the same time, the use of highly reactive, bioorthogonal functional groups (inverse electron demand Diels-Alder cycloaddition) on the nanoparticles allows an efficient ligation after administration inside the body, i.e., in vivo.
Collapse
Affiliation(s)
- Rudolf Zentel
- Department of Chemistry, Universität Mainz, Duesbergweg 10-14, D-55099 Mainz, Germany
| |
Collapse
|
17
|
Chauhan P, V R, Kumar M, Molla R, Mishra SD, Basa S, Rai V. Chemical technology principles for selective bioconjugation of proteins and antibodies. Chem Soc Rev 2024; 53:380-449. [PMID: 38095227 DOI: 10.1039/d3cs00715d] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Proteins are multifunctional large organic compounds that constitute an essential component of a living system. Hence, control over their bioconjugation impacts science at the chemistry-biology-medicine interface. A chemical toolbox for their precision engineering can boost healthcare and open a gateway for directed or precision therapeutics. Such a chemical toolbox remained elusive for a long time due to the complexity presented by the large pool of functional groups. The precise single-site modification of a protein requires a method to address a combination of selectivity attributes. This review focuses on guiding principles that can segregate them to simplify the task for a chemical method. Such a disintegration systematically employs a multi-step chemical transformation to deconvolute the selectivity challenges. It constitutes a disintegrate (DIN) theory that offers additional control parameters for tuning precision in protein bioconjugation. This review outlines the selectivity hurdles faced by chemical methods. It elaborates on the developments in the perspective of DIN theory to demonstrate simultaneous regulation of reactivity, chemoselectivity, site-selectivity, modularity, residue specificity, and protein specificity. It discusses the progress of such methods to construct protein and antibody conjugates for biologics, including antibody-fluorophore and antibody-drug conjugates (AFCs and ADCs). It also briefs how this knowledge can assist in developing small molecule-based covalent inhibitors. In the process, it highlights an opportunity for hypothesis-driven routes to accelerate discoveries of selective methods and establish new targetome in the precision engineering of proteins and antibodies.
Collapse
Affiliation(s)
- Preeti Chauhan
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| | - Ragendu V
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| | - Mohan Kumar
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| | - Rajib Molla
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| | - Surya Dev Mishra
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| | - Sneha Basa
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| | - Vishal Rai
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| |
Collapse
|
18
|
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
|
19
|
Morgan KA, Rudd SE, Noor A, Donnelly PS. Theranostic Nuclear Medicine with Gallium-68, Lutetium-177, Copper-64/67, Actinium-225, and Lead-212/203 Radionuclides. Chem Rev 2023; 123:12004-12035. [PMID: 37796539 DOI: 10.1021/acs.chemrev.3c00456] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2023]
Abstract
Molecular changes in malignant tissue can lead to an increase in the expression levels of various proteins or receptors that can be used to target the disease. In oncology, diagnostic imaging and radiotherapy of tumors is possible by attaching an appropriate radionuclide to molecules that selectively bind to these target proteins. The term "theranostics" describes the use of a diagnostic tool to predict the efficacy of a therapeutic option. Molecules radiolabeled with γ-emitting or β+-emitting radionuclides can be used for diagnostic imaging using single photon emission computed tomography or positron emission tomography. Radionuclide therapy of disease sites is possible with either α-, β-, or Auger-emitting radionuclides that induce irreversible damage to DNA. This Focus Review centers on the chemistry of theranostic approaches using metal radionuclides for imaging and therapy. The use of tracers that contain β+-emitting gallium-68 and β-emitting lutetium-177 will be discussed in the context of agents in clinical use for the diagnostic imaging and therapy of neuroendocrine tumors and prostate cancer. A particular emphasis is then placed on the chemistry involved in the development of theranostic approaches that use copper-64 for imaging and copper-67 for therapy with functionalized sarcophagine cage amine ligands. Targeted therapy with radionuclides that emit α particles has potential to be of particular use in late-stage disease where there are limited options, and the role of actinium-225 and lead-212 in this area is also discussed. Finally, we highlight the challenges that impede further adoption of radiotheranostic concepts while highlighting exciting opportunities and prospects.
Collapse
Affiliation(s)
- Katherine A Morgan
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| | - Stacey E Rudd
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| | - Asif Noor
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| |
Collapse
|
20
|
Edelmann MR, Bredack C, Belli S, Mohr P, Imhoff MP, Reggiani F, Kusznir EA, Rufer AC, Holt DP, Valentine H, Wong DF, Dannals RF, Honer M, Gobbi LC. Evaluation of Tetrazine Tracers for Pretargeted Imaging within the Central Nervous System. Bioconjug Chem 2023; 34:1882-1893. [PMID: 37710950 DOI: 10.1021/acs.bioconjchem.3c00385] [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: 09/16/2023]
Abstract
The pretargeting approach separates the biological half-life of an antibody from the physical half-life of the radioisotope label, providing a strategy for reducing the radiation burden. A widely explored pretargeting approach makes use of the bioorthogonal click reaction between tetrazines (Tzs) and trans-cyclooctenes (TCOs), combining the targeting specificity of monoclonal antibodies (mAbs) with the rapid clearance and precise reaction of Tzs and TCOs. Such a strategy can allow for the targeting and imaging (e.g., by positron emission tomography (PET)) of molecular markers, which cannot be addressed by solely relying on small molecules. Tz derivatives that undergo inverse electron-demand Diels-Alder (IEDDA) reactions with an antibody bearing TCO moieties have been investigated. This study describes the synthesis and characterization of 11 cold Tz imaging agent candidates. These molecules have the potential to be radiolabeled with 18F or 3H, and with the former label, they could be of use as imaging tracers for positron emission tomography studies. Selection was made using a multiparameter optimization score for the central nervous system (CNS) PET tracers. Novel tetrazines were tested for their pH-dependent chemical stability. Those which turned out to be stable in a pH range of 6.5-8 were further characterized in in vitro assays with regard to their passive permeability, microsomal stability, and P-glycoprotein transport. Furthermore, selected Tzs were examined for their systemic clearance and CNS penetration in a single-dose pharmacokinetic study in rats. Two tetrazines were successfully labeled with 18F, one of which showed brain penetration in a biodistribution study in mice. Another Tz was successfully tritium-labeled and used to demonstrate a bioorthogonal click reaction on a TCO-modified antibody. As a result, we identified one Tz as a potential fluorine-18-labeled CNS-PET agent and a second as a 3H-radioligand for an IEDDA-based reaction with a modified brain-penetrating antibody.
Collapse
Affiliation(s)
- Martin R Edelmann
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Therapeutic Modalities, Small Molecule Research, Isotope Synthesis, F. Hoffmann-La Roche Ltd, Basel CH-4070, Switzerland
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, U.K
| | - Christoph Bredack
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Neuroscience and Rare Diseases, Discovery & Translational Medicine Area, Biomarker and Translational Technologies, F. Hoffmann-La Roche Ltd, Basel CH-4070, Switzerland
| | - Sara Belli
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Pharmaceutical Science, F. Hoffmann-La Roche Ltd, Basel CH-4070, Switzerland
| | - Peter Mohr
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Therapeutic Modalities, Small Molecule Research, Medicinal Chemistry, F. Hoffmann-La Roche Ltd, Basel CH-4070, Switzerland
| | - Marie-Paule Imhoff
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Therapeutic Modalities, Small Molecule Research, Medicinal Chemistry, F. Hoffmann-La Roche Ltd, Basel CH-4070, Switzerland
| | - Flore Reggiani
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Therapeutic Modalities, Small Molecule Research, Medicinal Chemistry, F. Hoffmann-La Roche Ltd, Basel CH-4070, Switzerland
| | - Eric A Kusznir
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Therapeutic Modalities, Small Molecule Research, Lead Discovery, F. Hoffmann-La Roche Ltd, Basel CH-4070, Switzerland
| | - Arne C Rufer
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Therapeutic Modalities, Small Molecule Research, Lead Discovery, F. Hoffmann-La Roche Ltd, Basel CH-4070, Switzerland
| | - Daniel P Holt
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
| | - Heather Valentine
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
- Section of High Resolution Brain PET, PET Center, Division of Nuclear Medicine, Russell H. Morgan Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
| | - Dean F Wong
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
- Section of High Resolution Brain PET, PET Center, Division of Nuclear Medicine, Russell H. Morgan Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
- Section of High Resolution Brain PET, PET Center, Division of Nuclear Medicine, Russell H. Morgan Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, 21218, United States
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
- ⧫Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
| | - Robert F Dannals
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
| | - Michael Honer
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Neuroscience and Rare Diseases, Discovery & Translational Medicine Area, Biomarker and Translational Technologies, F. Hoffmann-La Roche Ltd, Basel CH-4070, Switzerland
| | - Luca C Gobbi
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Therapeutic Modalities, Small Molecule Research, Medicinal Chemistry, F. Hoffmann-La Roche Ltd, Basel CH-4070, Switzerland
| |
Collapse
|
21
|
Yang J, Zhu B, Ran C. The Application of Bio-orthogonality for In Vivo Animal Imaging. CHEMICAL & BIOMEDICAL IMAGING 2023; 1:434-447. [PMID: 37655167 PMCID: PMC10466453 DOI: 10.1021/cbmi.3c00033] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 09/02/2023]
Abstract
The application of bio-orthogonality has greatly facilitated numerous aspects of biological studies in recent years. In particular, bio-orthogonal chemistry has transformed biological research, including in vitro conjugate chemistry, target identification, and biomedical imaging. In this review, we highlighted examples of bio-orthogonal in vivo imaging published in recent years. We grouped the references into two major categories: bio-orthogonal chemistry-related imaging and in vivo imaging with bio-orthogonal nonconjugated pairing. Lastly, we discussed the challenges and opportunities of bio-orthogonality for in vivo imaging.
Collapse
Affiliation(s)
- Jun Yang
- Athinoula
A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Room 2301, Building 149, Charlestown, Boston, Massachusetts 02129, United States
| | - Biyue Zhu
- Athinoula
A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Room 2301, Building 149, Charlestown, Boston, Massachusetts 02129, United States
| | - Chongzhao Ran
- Athinoula
A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Room 2301, Building 149, Charlestown, Boston, Massachusetts 02129, United States
| |
Collapse
|
22
|
Otaru S, Martinmäki T, Kuurne I, Paulus A, Helariutta K, Sarparanta M, Airaksinen AJ. Radiolabelling of peptides with tetrazine ligation based on the inverse electron-demand Diels-Alder reaction: rapid, catalyst-free and mild conversion of 1,4-dihydropyridazines to pyridazines. RSC Adv 2023; 13:22606-22615. [PMID: 37501774 PMCID: PMC10369045 DOI: 10.1039/d3ra02807k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/16/2023] [Indexed: 07/29/2023] Open
Abstract
Click chemistry reactions, such as the tetrazine ligation, based on the inverse-electron demand Diels-Alder (IEDDA), are chemoselective cycloaddition reactions widely used for chemical modifications and synthesis of biomolecule-based radiopharmaceuticals for positron emission tomography (PET). The reactions have potential also for pretargeted PET imaging. When used as a bioconjugation method in production of biomolecule-based radiopharmaceuticals, IEDDA-based tetrazine ligation has one significant drawback, namely the formation of a mixture comprising reduced metastable dihydropyridazines (DHPs) and oxidized cycloadducts. Conversion of the reduced DHPs to stable pyridazines requires oxidation, which is typically achieved by using oxidants or by photo-irradiated air-oxidation, both methods requiring added reagents or reaction times of several hours, not compatible with short-lived radionuclides. Here we report a mild, rapid, and catalyst-free conversion of the DHPs to pyridazines. In this study, a model peptide Tyr3-octreotide (TOC) was modified with polyethylene glycol (PEG) linkers and with trans-cyclooctenes (TCOs) for rapid IEDDA-mediated radiolabeling. Fluorine-18-labelled alkylammoniomethyltrifluoroborate ([18F]AmBF3) tetrazines were conjugated to the TCO-TOC analogs at room temperature for rapid synthesis of PET imaging agent candidates. The formed DHPs were successfully converted to the oxidized form, after heating the radiolabelled bioconjugates in aqueous solution (≥95% water) at 60 °C for a minimum of 10 minutes in the presence of air, resulting in one-pot back-to-back IEDDA reaction and DHP conversion. The water content of the reaction mixture was to be found critical for the coversion. Our finding offers a straightforward method for conversion of the metastable DHPs from the IEDDA-based tetrazine ligation to stable, oxidized pyridazines. The method is especially suitable for applications requiring rapid conversion.
Collapse
Affiliation(s)
- Sofia Otaru
- Department of Chemistry, Radiochemistry, University of Helsinki Finland
| | - Tatu Martinmäki
- Department of Chemistry, Radiochemistry, University of Helsinki Finland
| | - Iida Kuurne
- Department of Chemistry, Radiochemistry, University of Helsinki Finland
| | - Andreas Paulus
- Department of Chemistry, Radiochemistry, University of Helsinki Finland
| | | | - Mirkka Sarparanta
- Department of Chemistry, Radiochemistry, University of Helsinki Finland
| | - Anu J Airaksinen
- Department of Chemistry, Radiochemistry, University of Helsinki Finland
- Turku PET Centre, University of Turku Kiinamyllynkatu 4-8 FI-20520 Turku Finland
- Department of Chemistry, University of Turku Finland
| |
Collapse
|
23
|
Klauser PC, Chopra S, Cao L, Bobba KN, Yu B, Seo Y, Chan E, Flavell RR, Evans MJ, Wang L. Covalent Proteins as Targeted Radionuclide Therapies Enhance Antitumor Effects. ACS CENTRAL SCIENCE 2023; 9:1241-1251. [PMID: 37396859 PMCID: PMC10311652 DOI: 10.1021/acscentsci.3c00288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Indexed: 07/04/2023]
Abstract
Molecularly targeted radionuclide therapies (TRTs) struggle with balancing efficacy and safety, as current strategies to increase tumor absorption often alter drug pharmacokinetics to prolong circulation and normal tissue irradiation. Here we report the first covalent protein TRT, which, through reacting with the target irreversibly, increases radioactive dose to the tumor without altering the drug's pharmacokinetic profile or normal tissue biodistribution. Through genetic code expansion, we engineered a latent bioreactive amino acid into a nanobody, which binds to its target protein and forms a covalent linkage via the proximity-enabled reactivity, cross-linking the target irreversibly in vitro, on cancer cells, and on tumors in vivo. The radiolabeled covalent nanobody markedly increases radioisotope levels in tumors and extends tumor residence time while maintaining rapid systemic clearance. Furthermore, the covalent nanobody conjugated to the α-emitter actinium-225 inhibits tumor growth more effectively than the noncovalent nanobody without causing tissue toxicity. Shifting the protein-based TRT from noncovalent to covalent mode, this chemical strategy improves tumor responses to TRTs and can be readily scaled to diverse protein radiopharmaceuticals engaging broad tumor targets.
Collapse
Affiliation(s)
- Paul C. Klauser
- Department
of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158, United States
| | - Shalini Chopra
- Department
of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158, United States
- Department
of Radiology and Biomedical Imaging, University
of California San Francisco, San Francisco, California 94158, United States
| | - Li Cao
- Department
of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158, United States
| | - Kondapa Naidu Bobba
- Department
of Radiology and Biomedical Imaging, University
of California San Francisco, San Francisco, California 94158, United States
| | - Bingchen Yu
- Department
of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158, United States
| | - Youngho Seo
- Department
of Radiology and Biomedical Imaging, University
of California San Francisco, San Francisco, California 94158, United States
| | - Emily Chan
- Department
of Pathology, University of California San
Francisco, San Francisco, California 94158, United States
| | - Robert R. Flavell
- Department
of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158, United States
- Department
of Radiology and Biomedical Imaging, University
of California San Francisco, San Francisco, California 94158, United States
| | - Michael J. Evans
- Department
of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158, United States
- Department
of Radiology and Biomedical Imaging, University
of California San Francisco, San Francisco, California 94158, United States
| | - Lei Wang
- Department
of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158, United States
| |
Collapse
|
24
|
Chaney AM, Cropper HC, Jain P, Wilson E, Simonetta F, Johnson EM, Alam IS, Patterson ITJ, Swarovski M, Stevens MY, Wang Q, Azevedo C, Nagy SC, Ramos Benitez J, Deal EM, Vogel H, Andreasson KI, James ML. PET imaging of TREM1 identifies CNS-infiltrating myeloid cells in a mouse model of multiple sclerosis. Sci Transl Med 2023; 15:eabm6267. [PMID: 37379371 DOI: 10.1126/scitranslmed.abm6267] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 06/02/2023] [Indexed: 06/30/2023]
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the central nervous system (CNS) that causes substantial morbidity and diminished quality of life. Evidence highlights the central role of myeloid lineage cells in the initiation and progression of MS. However, existing imaging strategies for detecting myeloid cells in the CNS cannot distinguish between beneficial and harmful immune responses. Thus, imaging strategies that specifically identify myeloid cells and their activation states are critical for MS disease staging and monitoring of therapeutic responses. We hypothesized that positron emission tomography (PET) imaging of triggering receptor expressed on myeloid cells 1 (TREM1) could be used to monitor deleterious innate immune responses and disease progression in the experimental autoimmune encephalomyelitis (EAE) mouse model of MS. We first validated TREM1 as a specific marker of proinflammatory, CNS-infiltrating, peripheral myeloid cells in mice with EAE. We show that the 64Cu-radiolabeled TREM1 antibody-based PET tracer monitored active disease with 14- to 17-fold higher sensitivity than translocator protein 18 kDa (TSPO)-PET imaging, the established approach for detecting neuroinflammation in vivo. We illustrate the therapeutic potential of attenuating TREM1 signaling both genetically and pharmacologically in the EAE mice and show that TREM1-PET imaging detected responses to an FDA-approved MS therapy with siponimod (BAF312) in these animals. Last, we observed TREM1+ cells in clinical brain biopsy samples from two treatment-naïve patients with MS but not in healthy control brain tissue. Thus, TREM1-PET imaging has potential for aiding in the diagnosis of MS and monitoring of therapeutic responses to drug treatment.
Collapse
Affiliation(s)
- Aisling M Chaney
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Haley C Cropper
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Poorva Jain
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Edward Wilson
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA 94305, USA
| | - Federico Simonetta
- Division of Hematology, Department of Oncology, Geneva University Hospitals, Geneva 1205, Switzerland
- Translational Research Centre in Onco-Haematology, Faculty of Medicine, University of Geneva, Geneva 1205, Switzerland
| | - Emily M Johnson
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Israt S Alam
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Ian T J Patterson
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Michelle Swarovski
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA 94305, USA
| | - Marc Y Stevens
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Qian Wang
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA 94305, USA
| | - Carmen Azevedo
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Sydney C Nagy
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Javier Ramos Benitez
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA 94305, USA
| | - Emily M Deal
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Hannes Vogel
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Katrin I Andreasson
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Michelle L James
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
25
|
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
|
26
|
van Dijk B, Hooning van Duyvenbode JFF, de Vor L, Nurmohamed FRHA, Lam MGEH, Poot AJ, Ramakers RM, Koustoulidou S, Beekman FJ, van Strijp J, Rooijakkers SHM, Dadachova E, Vogely HC, Weinans H, van der Wal BCH. Evaluating the Targeting of a Staphylococcus-aureus-Infected Implant with a Radiolabeled Antibody In Vivo. Int J Mol Sci 2023; 24:ijms24054374. [PMID: 36901805 PMCID: PMC10002501 DOI: 10.3390/ijms24054374] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/06/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
Implant infections caused by Staphylococcus aureus are difficult to treat due to biofilm formation, which complicates surgical and antibiotic treatment. We introduce an alternative approach using monoclonal antibodies (mAbs) targeting S. aureus and provide evidence of the specificity and biodistribution of S.-aureus-targeting antibodies in a mouse implant infection model. The monoclonal antibody 4497-IgG1 targeting wall teichoic acid in S. aureus was labeled with indium-111 using CHX-A"-DTPA as a chelator. Single Photon Emission Computed Tomography/computed tomographyscans were performed at 24, 72 and 120 h after administration of the 111In-4497 mAb in Balb/cAnNCrl mice with a subcutaneous implant that was pre-colonized with S. aureus biofilm. The biodistribution of this labelled antibody over various organs was visualized and quantified using SPECT/CT imaging, and was compared to the uptake at the target tissue with the implanted infection. Uptake of the 111In-4497 mAbs at the infected implant gradually increased from 8.34 %ID/cm3 at 24 h to 9.22 %ID/cm3 at 120 h. Uptake at the heart/blood pool decreased over time from 11.60 to 7.58 %ID/cm3, whereas the uptake in the other organs decreased from 7.26 to less than 4.66 %ID/cm3 at 120 h. The effective half-life of 111In-4497 mAbs was determined to be 59 h. In conclusion, 111In-4497 mAbs were found to specifically detect S. aureus and its biofilm with excellent and prolonged accumulation at the site of the colonized implant. Therefore, it has the potential to serve as a drug delivery system for the diagnostic and bactericidal treatment of biofilm.
Collapse
Affiliation(s)
- Bruce van Dijk
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Correspondence: ; Tel.: +31-88-75-569-71
| | | | - Lisanne de Vor
- Department of Medical Microbiology, University Medical Centre Utrecht, 3584 CX Utrecht, The Netherlands
| | | | - Marnix G. E. H. Lam
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Alex J. Poot
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Ruud M. Ramakers
- MILabs B.V., 3584 CX Utrecht, The Netherlands
- Department of Radiation Science and Technology, Delft University of Technology, 2628 CD Delft, The Netherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center, 3584 CX Utrecht, The Netherlands
| | - Sofia Koustoulidou
- MILabs B.V., 3584 CX Utrecht, The Netherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center, 3584 CX Utrecht, The Netherlands
| | - Freek J. Beekman
- MILabs B.V., 3584 CX Utrecht, The Netherlands
- Department of Radiation Science and Technology, Delft University of Technology, 2628 CD Delft, The Netherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center, 3584 CX Utrecht, The Netherlands
| | - Jos van Strijp
- Department of Medical Microbiology, University Medical Centre Utrecht, 3584 CX Utrecht, The Netherlands
| | - Suzan H. M. Rooijakkers
- Department of Medical Microbiology, University Medical Centre Utrecht, 3584 CX Utrecht, The Netherlands
| | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5A8, Canada
| | - H. Charles Vogely
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Harrie Weinans
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Department of BioMechanical Engineering, Delft University of Technology, 2628 CD Delft, The Netherlands
| | - Bart C. H. van der Wal
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
27
|
Bidesi N, Shalgunov V, Battisti UM, Hvass L, Jørgensen JT, Poulie CBM, Jensen AI, Kjaer A, Herth MM. Synthesis and radiolabeling of a polar [ 125 I]I-1,2,4,5-tetrazine. J Labelled Comp Radiopharm 2023; 66:22-30. [PMID: 36539610 PMCID: PMC10107300 DOI: 10.1002/jlcr.4009] [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: 08/22/2022] [Revised: 10/13/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
Pretargeting imaging has gained a lot of prominence, due to its excellent bioorthogonality and improved imaging contrast compared to conventional imaging. A new iodo tetrazine (Tz) derivative has been synthesized and further developed into the corresponding iodine-125 (125 I) analog (12), via the trimethylstannane precursor. Radiolabeling with either N-chlorosuccinimide or chloramine-T, in either MeCN or MeOH proceeded with a radiochemical conversion (RCC) of >80%. Subsequent deprotection only proved successful, among the tested conditions, when the radiolabeled Tz was stirred in 6-M HCl(aq.) at 60°C for 2.5 h. To the best of our knowledge, this is the first H-tetrazine labeled with iodine. In vivo investigations on the pretargeting ability of 12 are currently under way.
Collapse
Affiliation(s)
- Natasha Bidesi
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Copenhagen, Denmark
| | - Umberto Maria Battisti
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars Hvass
- Department of Clinical Physiology, Nuclear Medicine and PET & Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Tranekjaer Jørgensen
- Department of Clinical Physiology, Nuclear Medicine and PET & Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christian B M Poulie
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andreas I Jensen
- Center for Nuclear Technologies (DTU Nutech), Technical University of Denmark (DTU), Roskilde, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine and PET & Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Matthias M Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
28
|
Cook BE, Archbold J, Nasr K, Girmay S, Goldstein SI, Li P, Dandapani S, Genung NE, Tang SP, McClusky S, Plisson C, Afetian ME, Dwyer CA, Fazio M, Drury WJ, Rigo F, Martarello L, Kaliszczak M. Non-invasive Imaging of Antisense Oligonucleotides in the Brain via In Vivo Click Chemistry. Mol Imaging Biol 2022; 24:940-949. [PMID: 35655109 DOI: 10.1007/s11307-022-01744-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/10/2022] [Accepted: 05/22/2022] [Indexed: 12/29/2022]
Abstract
PURPOSE The treatment of complex neurological diseases often requires the administration of large therapeutic drugs, such as antisense oligonucleotide (ASO), by lumbar puncture into the intrathecal space in order to bypass the blood-brain barrier. Despite the growing number of ASOs in clinical development, there are still uncertainties regarding their dosing, primarily around their distribution and kinetics in the brain following intrathecal injection. The challenge of taking measurements within the delicate structures of the central nervous system (CNS) necessitates the use of non-invasive nuclear imaging, such as positron emission tomography (PET). Herein, an emergent strategy known as "pretargeted imaging" is applied to image the distribution of an ASO in the brain by developing a novel PET tracer, [18F]F-537-Tz. This tracer is able to undergo an in vivo "click" reaction, covalently binding to a trans-cyclooctene conjugated ASO. PROCEDURES A novel small molecule tracer for pretargeted PET imaging of ASOs in the CNS is developed and tested in a series of in vitro and in vivo experiments, including biodistribution in rats and non-human primates. RESULTS In vitro data and extensive in vivo rat data demonstrated delivery of the tracer to the CNS, and its successful ligation to its ASO target in the brain. In an NHP study, the slow tracer kinetics did not allow for specific binding to be determined by PET. CONCLUSION A CNS-penetrant radioligand for pretargeted imaging was successfully demonstrated in a proof-of-concept study in rats, laying the groundwork for further optimization.
Collapse
Affiliation(s)
| | | | - Khaled Nasr
- Invicro, A Konica Minolta Company, Boston, MA, 02210, USA
| | | | | | - Pei Li
- , Biogen, Cambridge, MA, 02142, USA
| | | | | | - Sac-Pham Tang
- Invicro, A Konica Minolta Company, Boston, MA, 02210, USA
| | | | | | | | | | | | | | - Frank Rigo
- Ionis Pharmaceuticals Inc, Carlsbad, CA, 92010, USA
| | | | | |
Collapse
|
29
|
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
|
30
|
Scinto SL, Reagle TR, Fox JM. Affinity Bioorthogonal Chemistry (ABC) Tags for Site-Selective Conjugation, On-Resin Protein-Protein Coupling, and Purification of Protein Conjugates. Angew Chem Int Ed Engl 2022; 61:e202207661. [PMID: 36058881 PMCID: PMC10029600 DOI: 10.1002/anie.202207661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Indexed: 11/12/2022]
Abstract
The site-selective functionalization of proteins has broad application in chemical biology, but can be limited when mixtures result from incomplete conversion or the formation of protein containing side products. It is shown here that when proteins are covalently tagged with pyridyl-tetrazines, the nickel-iminodiacetate (Ni-IDA) resins commonly used for His-tags can be directly used for protein affinity purification. These Affinity Bioorthogonal Chemistry (ABC) tags serve a dual role by enabling affinity-based protein purification while maintaining rapid kinetics in bioorthogonal reactions. ABC-tagging works with a range of site-selective bioconjugation methods with proteins tagged at the C-terminus, N-terminus or at internal positions. ABC-tagged proteins can also be purified from complex mixtures including cell lysate. The combination of site-selective conjugation and clean-up with ABC-tagged proteins also allows for facile on-resin reactions to provide protein-protein conjugates.
Collapse
Affiliation(s)
- Samuel L Scinto
- Department of Chemistry and Biochemistry, University of Delaware, Ammon Pinizzotto Biopharmaceutical Innovation Center, Newark, DE 19713, USA
| | - Tyler R Reagle
- Department of Chemistry and Biochemistry, University of Delaware, Ammon Pinizzotto Biopharmaceutical Innovation Center, Newark, DE 19713, USA
| | - Joseph M Fox
- Department of Chemistry and Biochemistry, University of Delaware, Ammon Pinizzotto Biopharmaceutical Innovation Center, Newark, DE 19713, USA
| |
Collapse
|
31
|
Bhise A, Park H, Lee W, Sarkar S, Ha YS, Rajkumar S, Nam B, Lim JE, Huynh PT, Lee K, Son JY, Kim JY, Lee KC, Yoo J. Preclinical Evaluation of hnRNPA2B1 Antibody in Human Triple-Negative Breast Cancer MDA-MB-231 Cells via PET Imaging. Pharmaceutics 2022; 14:pharmaceutics14081677. [PMID: 36015303 PMCID: PMC9415040 DOI: 10.3390/pharmaceutics14081677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/27/2022] [Accepted: 08/09/2022] [Indexed: 11/25/2022] Open
Abstract
Triple-negative breast cancer (TNBC) does not express estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2. Because TNBC lacks the expression of commonly targeted receptors, it is challenging to develop a new imaging agent for this cancer subtype. Heterogeneous nuclear ribonucleoproteins (hnRNPs) are RNA–protein complexes that have been linked to tumor development and progression. Considering the high expression of hnRNPA2B1, an hnRNP subtype, in TNBC MDA-MB-231 cells, this study aimed to develop a novel hnRNPA2B1 antibody-based nuclear imaging agent. The hnRNPA2B1-specific antibody was radiolabeled with 64Cu and evaluated in vitro and in vivo. The trans-cyclooctene (TCO) was functionalized on the antibody to obtain hnRNP-PEG4-TCO and reactive tetrazine (Tz) on the ultrastable bifunctional chelator PCB-TE2A-alkyne to yield PCB-TE2A-Tz for the inverse electron demand Diels–Alder reaction. The 64Cu-radiolabeled antibody was administered and imaged at 1–18 h time points for conventional imaging. Alternatively, the unlabeled antibody conjugate was administered, and 48 h later radiolabeled 64Cu-PCB-TE2A-Tz was administered to the same mice for the pretargeting strategy and imaged at the same time intervals for direct comparison. The tumor was successfully visualized in both strategies, and comparatively, pretargeting showed superior results. The 64Cu-PCB-TE2A-Tz was successfully clicked at the tumor site with hnRNP-PEG4-TCO and the non-clicked were concurrently eliminated. This led to increase the tumor uptake with extremely high tumor-to-background ratio manifested by positron emission tomography (PET) imaging and biodistribution studies.
Collapse
Affiliation(s)
- Abhinav Bhise
- Department of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Hyun Park
- Division of Applied RI, Korea Institute of Radiological and Medical Science, Seoul 01812, Korea
| | - Woonghee Lee
- Department of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Swarbhanu Sarkar
- Department of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Yeong Su Ha
- Department of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Subramani Rajkumar
- Department of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Bora Nam
- Department of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Jeong Eun Lim
- Department of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Phuong Tu Huynh
- Department of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Kiwoong Lee
- Department of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Ji-Yoon Son
- Department of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Jung Young Kim
- Division of Applied RI, Korea Institute of Radiological and Medical Science, Seoul 01812, Korea
| | - Kyo Chul Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Science, Seoul 01812, Korea
| | - Jeongsoo Yoo
- Department of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Correspondence: ; Tel.: +82-53-420-4947
| |
Collapse
|
32
|
Bodei L, Herrmann K, Schöder H, Scott AM, Lewis JS. Radiotheranostics in oncology: current challenges and emerging opportunities. Nat Rev Clin Oncol 2022; 19:534-550. [PMID: 35725926 PMCID: PMC10585450 DOI: 10.1038/s41571-022-00652-y] [Citation(s) in RCA: 179] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2022] [Indexed: 12/20/2022]
Abstract
Structural imaging remains an essential component of diagnosis, staging and response assessment in patients with cancer; however, as clinicians increasingly seek to noninvasively investigate tumour phenotypes and evaluate functional and molecular responses to therapy, theranostics - the combination of diagnostic imaging with targeted therapy - is becoming more widely implemented. The field of radiotheranostics, which is the focus of this Review, combines molecular imaging (primarily PET and SPECT) with targeted radionuclide therapy, which involves the use of small molecules, peptides and/or antibodies as carriers for therapeutic radionuclides, typically those emitting α-, β- or auger-radiation. The exponential, global expansion of radiotheranostics in oncology stems from its potential to target and eliminate tumour cells with minimal adverse effects, owing to a mechanism of action that differs distinctly from that of most other systemic therapies. Currently, an enormous opportunity exists to expand the number of patients who can benefit from this technology, to address the urgent needs of many thousands of patients across the world. In this Review, we describe the clinical experience with established radiotheranostics as well as novel areas of research and various barriers to progress.
Collapse
Affiliation(s)
- Lisa Bodei
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Weill Cornell Medical School, New York, NY, USA
| | - Ken Herrmann
- German Cancer Consortium, University Hospital Essen, Essen, Germany
- Department of Nuclear Medicine, University of Duisburg-Essen, University Hospital Essen, Essen, Germany
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Weill Cornell Medical School, New York, NY, USA
| | - Andrew M Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiology, Weill Cornell Medical School, New York, NY, USA.
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Pharmacology, Weill Cornell Medical School, New York, NY, USA.
| |
Collapse
|
33
|
Caers J, Duray E, Vrancken L, Marcion G, Bocuzzi V, De Veirman K, Krasniqi A, Lejeune M, Withofs N, Devoogdt N, Dumoulin M, Karlström AE, D’Huyvetter M. Radiotheranostic Agents in Hematological Malignancies. Front Immunol 2022; 13:911080. [PMID: 35865548 PMCID: PMC9294596 DOI: 10.3389/fimmu.2022.911080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/06/2022] [Indexed: 12/23/2022] Open
Abstract
Radioimmunotherapy (RIT) is a cancer treatment that combines radiation therapy with tumor-directed monoclonal antibodies (Abs). Although RIT had been introduced for the treatment of CD20 positive non-Hodgkin lymphoma decades ago, it never found a broad clinical application. In recent years, researchers have developed theranostic agents based on Ab fragments or small Ab mimetics such as peptides, affibodies or single-chain Abs with improved tumor-targeting capacities. Theranostics combine diagnostic and therapeutic capabilities into a single pharmaceutical agent; this dual application can be easily achieved after conjugation to radionuclides. The past decade has seen a trend to increased specificity, fastened pharmacokinetics, and personalized medicine. In this review, we discuss the different strategies introduced for the noninvasive detection and treatment of hematological malignancies by radiopharmaceuticals. We also discuss the future applications of these radiotheranostic agents.
Collapse
Affiliation(s)
- Jo Caers
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
- Department of Hematology, CHU de Liège, Liège, Belgium
- *Correspondence: Jo Caers,
| | - Elodie Duray
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
- Centre for Protein Engineering, Inbios, University of Liège, Liège, Belgium
| | - Louise Vrancken
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
- Department of Hematology, CHU de Liège, Liège, Belgium
| | - Guillaume Marcion
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
| | - Valentina Bocuzzi
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
| | - Kim De Veirman
- Department of Hematology and Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ahmet Krasniqi
- Laboratory of In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium
| | - Margaux Lejeune
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
| | - Nadia Withofs
- Department of Nuclear Medicine, CHU de Liège, Liège, Belgium
| | - Nick Devoogdt
- Laboratory of In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium
| | - Mireille Dumoulin
- Centre for Protein Engineering, Inbios, University of Liège, Liège, Belgium
| | - Amelie Eriksson Karlström
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Matthias D’Huyvetter
- Laboratory of In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
34
|
Manafi-Farid R, Ataeinia B, Ranjbar S, Jamshidi Araghi Z, Moradi MM, Pirich C, Beheshti M. ImmunoPET: Antibody-Based PET Imaging in Solid Tumors. Front Med (Lausanne) 2022; 9:916693. [PMID: 35836956 PMCID: PMC9273828 DOI: 10.3389/fmed.2022.916693] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/24/2022] [Indexed: 12/13/2022] Open
Abstract
Immuno-positron emission tomography (immunoPET) is a molecular imaging modality combining the high sensitivity of PET with the specific targeting ability of monoclonal antibodies. Various radioimmunotracers have been successfully developed to target a broad spectrum of molecules expressed by malignant cells or tumor microenvironments. Only a few are translated into clinical studies and barely into clinical practices. Some drawbacks include slow radioimmunotracer kinetics, high physiologic uptake in lymphoid organs, and heterogeneous activity in tumoral lesions. Measures are taken to overcome the disadvantages, and new tracers are being developed. In this review, we aim to mention the fundamental components of immunoPET imaging, explore the groundbreaking success achieved using this new technique, and review different radioimmunotracers employed in various solid tumors to elaborate on this relatively new imaging modality.
Collapse
Affiliation(s)
- Reyhaneh Manafi-Farid
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahar Ataeinia
- Department of Radiology, Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Shaghayegh Ranjbar
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Zahra Jamshidi Araghi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mobin Moradi
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Christian Pirich
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Mohsen Beheshti
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
35
|
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
|
36
|
Matiz CA, Delaney S, Cook BE, Genady AR, Hoerres R, Kuchuk M, Makris G, Valliant JF, Sadeghi S, Lewis JS, Hennkens HM, Bryan JN, Zeglis BM. Pretargeted PET of Osteodestructive Lesions in Dogs. Mol Pharm 2022; 19:3153-3162. [DOI: 10.1021/acs.molpharmaceut.2c00220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Charles A. Matiz
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri 65211, United States
| | - Samantha Delaney
- Department of Chemistry, Hunter College, City University of New York, New York, New York 10065, United States
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Brendon E. Cook
- Department of Chemistry, Hunter College, City University of New York, New York, New York 10065, United States
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
| | - Afaf R. Genady
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Rebecca Hoerres
- Department of Chemistry and Research Reactor, University of Missouri, Columbia, Missouri 65211, United States
| | - Marina Kuchuk
- Department of Chemistry and Research Reactor, University of Missouri, Columbia, Missouri 65211, United States
| | - Georgios Makris
- Department of Chemistry and Research Reactor, University of Missouri, Columbia, Missouri 65211, United States
| | - John F. Valliant
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Saman Sadeghi
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Jason S. Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Department of Pharmacology, Weill Cornell Medical College, New York, New York 10021, United States
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Department of Radiology, Weill Cornell Medical College, New York, New York 10021, United States
| | - Heather M. Hennkens
- Department of Chemistry and Research Reactor, University of Missouri, Columbia, Missouri 65211, United States
| | - Jeffrey N. Bryan
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri 65211, United States
| | - Brian M. Zeglis
- Department of Chemistry, Hunter College, City University of New York, New York, New York 10065, United States
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
- Department of Radiology, Weill Cornell Medical College, New York, New York 10021, United States
| |
Collapse
|
37
|
Svatunek D, Wilkovitsch M, Hartmann L, Houk KN, Mikula H. Uncovering the Key Role of Distortion in Bioorthogonal Tetrazine Tools That Defy the Reactivity/Stability Trade-Off. J Am Chem Soc 2022; 144:8171-8177. [PMID: 35500228 PMCID: PMC9100665 DOI: 10.1021/jacs.2c01056] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
The tetrazine/trans-cyclooctene ligation stands
out from the bioorthogonal toolbox due to its exceptional reaction
kinetics, enabling multiple molecular technologies in vitro and in
living systems. Highly reactive 2-pyridyl-substituted tetrazines have
become state of the art for time-critical processes and selective
reactions at very low concentrations. It is widely accepted that the
enhanced reactivity of these chemical tools is attributed to the electron-withdrawing
effect of the heteroaryl substituent. In contrast, we show that the
observed reaction rates are way too high to be explained on this basis.
Computational investigation of this phenomenon revealed that distortion
of the tetrazine caused by intramolecular repulsive N–N interaction
plays a key role in accelerating the cycloaddition step. We show that
the limited stability of tetrazines in biological media strongly correlates
with the electron-withdrawing effect of the substituent, while intramolecular
repulsion increases the reactivity without reducing the stability.
These fundamental insights reveal thus far overlooked mechanistic
aspects that govern the reactivity/stability trade-off for tetrazines
in physiologically relevant environments, thereby providing a new
strategy that may facilitate the rational design of these bioorthogonal
tools.
Collapse
Affiliation(s)
- Dennis Svatunek
- Institute of Applied Synthetic Chemistry, TU Wien 1060 Vienna, Austria
| | | | - Lea Hartmann
- Institute of Applied Synthetic Chemistry, TU Wien 1060 Vienna, Austria
| | - K N Houk
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles 90095, United States
| | - Hannes Mikula
- Institute of Applied Synthetic Chemistry, TU Wien 1060 Vienna, Austria
| |
Collapse
|
38
|
Improved Characteristics of RANKL Immuno-PET Imaging Using Radiolabeled Antibody Fab Fragments. Pharmaceutics 2022; 14:pharmaceutics14050939. [PMID: 35631525 PMCID: PMC9147590 DOI: 10.3390/pharmaceutics14050939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/15/2022] [Accepted: 04/20/2022] [Indexed: 01/25/2023] Open
Abstract
Purpose: RANKL expression in the tumor microenvironment has been identified as a biomarker of immune suppression, negating the effect of some cancer immunotherapies. Previously we had developed a radiotracer based on the FDA-approved RANKL-specific antibody denosumab, [89Zr]Zr-DFO-denosumab, enabling successful immuno-PET imaging. Radiolabeled denosumab, however, showed long blood circulation and delayed tumor uptake, potentially limiting its applications. Here we aimed to develop a smaller radiolabeled denosumab fragment, [64Cu]Cu-NOTA-denos-Fab, that would ideally show faster tumor accumulation and better diffusion into the tumor for the visualization of RANKL. Experimental design: Fab fragments were prepared from denosumab using papain and conjugated to a NOTA chelator for radiolabeling with 64Cu. The bioconjugates were characterized in vitro using SDS-PAGE analysis, and the binding affinity was assessed using a radiotracer cell binding assay. Small animal PET imaging evaluated tumor targeting and biodistribution in transduced RANKL-ME-180 xenografts. Results: The radiolabeling yield of [64Cu]Cu-NOTA-denos-Fab was 58 ± 9.2%, with a specific activity of 0.79 ± 0.11 MBq/µg (n = 3). A radiotracer binding assay proved specific targeting of RANKL in vitro. PET imaging showed fast blood clearance and high tumor accumulation as early as 1 h p.i. (2.14 ± 0.21% ID/mL), which peaked at 5 h p.i. (2.72 ± 0.61% ID/mL). In contrast, [64Cu]Cu-NOTA-denosumab reached its highest tumor uptake at 24 h p.i. (6.88 ± 1.12% ID/mL). [64Cu]Cu-NOTA-denos-Fab specifically targeted human RANKL in transduced ME-180 xenografts compared with the blocking group and negative ME-180 xenograft model. Histological analysis confirmed RANKL expression in RANKL-ME-180 xenografts. Conclusions: Here, we report on a novel RANKL PET imaging agent, [64Cu]Cu-NOTA-denos-Fab, that allows for fast tumor imaging with improved imaging contrast when compared with its antibody counterpart, showing promise as a potential PET RANKL imaging tool for future clinical applications.
Collapse
|
39
|
Lumen D, Vugts D, Chomet M, Imlimthan S, Sarparanta M, Vos R, Schreurs M, Verlaan M, Lang PA, Hippeläinen E, Beaino W, Windhorst AD, Airaksinen AJ. Pretargeted PET Imaging with a TCO-Conjugated Anti-CD44v6 Chimeric mAb U36 and [ 89Zr]Zr-DFO-PEG 5-Tz. Bioconjug Chem 2022; 33:956-968. [PMID: 35442642 PMCID: PMC9121349 DOI: 10.1021/acs.bioconjchem.2c00164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
The recent advances
in the production of engineered antibodies
have facilitated the development and application of tailored, target-specific
antibodies. Positron emission tomography (PET) of these antibody-based
drug candidates can help to better understand their in vivo behavior. In this study, we report an in vivo proof-of-concept
pretargeted immuno-PET study where we compare a pretargeting vs targeted
approach using a new 89Zr-labeled tetrazine as a bio-orthogonal
ligand in an inverse electron demand Diels–Alder (IEDDA) in vivo click reaction. A CD44v6-selective chimeric monoclonal
U36 was selected as the targeting antibody because it has potential
in immuno-PET imaging of head-and-neck squamous cell carcinoma (HNSCC).
Zirconium-89 (t1/2 = 78.41 h) was selected
as the radionuclide of choice to be able to make a head-to-head comparison
of the pretargeted and targeted approaches. [89Zr]Zr-DFO-PEG5-Tz ([89Zr]Zr-3) was synthesized and
used in pretargeted PET imaging of HNSCC xenografts (VU-SCC-OE) at
24 and 48 h after administration of a trans-cyclooctene
(TCO)-functionalized U36. The pretargeted approach resulted in lower
absolute tumor uptake than the targeted approach (1.5 ± 0.2 vs
17.1 ± 3.0% ID/g at 72 h p.i. U36) but with comparable tumor-to-non-target
tissue ratios and significantly lower absorbed doses. In conclusion,
anti-CD44v6 monoclonal antibody U36 was successfully used for 89Zr-immuno-PET imaging of HNSCC xenograft tumors using both
a targeted and pretargeted approach. The results not only support
the utility of the pretargeted approach in immuno-PET imaging but
also demonstrate the challenges in achieving optimal in vivo IEDDA reaction efficiencies in relation to antibody pharmacokinetics.
Collapse
Affiliation(s)
- Dave Lumen
- Department of Chemistry, Radiochemistry, University of Helsinki, FI-00014 Helsinki, Finland
| | - Danielle Vugts
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Marion Chomet
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Surachet Imlimthan
- Department of Chemistry, Radiochemistry, University of Helsinki, FI-00014 Helsinki, Finland
| | - Mirkka Sarparanta
- Department of Chemistry, Radiochemistry, University of Helsinki, FI-00014 Helsinki, Finland
| | - Ricardo Vos
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Maxime Schreurs
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Mariska Verlaan
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Pauline A Lang
- Department of Chemistry, Radiochemistry, University of Helsinki, FI-00014 Helsinki, Finland
| | - Eero Hippeläinen
- HUS Medical Imaging Center, Clinical Physiology and Nuclear Medicine, University of Helsinki and Helsinki University Hospital, 00029 HUS Helsinki, Finland
| | - Wissam Beaino
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Albert D Windhorst
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Anu J Airaksinen
- Department of Chemistry, Radiochemistry, University of Helsinki, FI-00014 Helsinki, Finland.,Turku PET Centre, Department of Chemistry, University of Turku, 20520 Turku, Finland
| |
Collapse
|
40
|
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
|
41
|
Kujawski M, Li L, Li H, Yazaki PJ, Swiderski P, Shively JE. T-cell surface generation of dual bivalent, bispecific T-cell engaging, RNA duplex cross-linked antibodies (dbBiTERs) for re-directed tumor cell lysis. Biotechnol J 2022; 17:e2100389. [PMID: 34773368 PMCID: PMC9177045 DOI: 10.1002/biot.202100389] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/06/2021] [Accepted: 11/09/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Genetic engineered Bispecific T-cell engagers (BiTEs) generate potent cytotoxic effects. METHODS Alternately, click chemistry engineered, dual specific bivalent Bispecific T-cell engaging antibodies (dbBiTEs) on T-cell surfaces can be generated from parent monoclonal antibodies. RESULTS We show the formation of dbBiTEs on the surface of T-cells along with the introduction of complementary 2'-OMe RNA 32-mer oligonucleotides allowing duplex formation between antibodies, designated as dbBiTERs. dbBiTERs generated in solution from anti-CEA and anti-CD3 OKT3 antibodies retained specific binding to CEA positive versus CEA negative cancer cells and to CD3 positive T-cells comparable to dbBiTEs. When T-cells were precoated with dbBiTEs or dbBiTERs and mixed with CEA positive versus CEA negative cancer cells, similar dose dependent and specific cytotoxicity were observed in redirected cell lysis assays. On-cell generated dbBiTERs exerted potent cytotoxic responses against CEA positive targets and were localized at the cell surface by immuno-gold EM. In addition, we demonstrate that target and T-cells, each coated separately with complementary 2'OMe-RNA-linked antibodies can be cross-linked by RNA duplex formation in vitro to generate redirected cell lysis. CONCLUSION The facile generation of dbBiTERs with specific cytolytic activity from intact antibodies and their generation on-cell offers a new avenue for antigen specific T-cell therapy.
Collapse
Affiliation(s)
- Maciej Kujawski
- Department of Immunology and Theranostics, City of Hope, Duarte, California, USA
| | - Lin Li
- Department of Immunology and Theranostics, City of Hope, Duarte, California, USA
| | - Harry Li
- Department of Immunology and Theranostics, City of Hope, Duarte, California, USA
| | - Paul J. Yazaki
- Department of Immunology and Theranostics, City of Hope, Duarte, California, USA
| | - Piotr Swiderski
- Shared Resources-DNA/RNA/Peptide, City of Hope, Duarte, California, USA
| | - John E. Shively
- Department of Immunology and Theranostics, City of Hope, Duarte, California, USA
| |
Collapse
|
42
|
Engudar G, Rodríguez-Rodríguez C, Mishra NK, Bergamo M, Amouroux G, Jensen KJ, Saatchi K, Häfeli UO. Metal-ion coordinated self-assembly of human insulin directs kinetics of insulin release as determined by preclinical SPECT/CT imaging. J Control Release 2022; 343:347-360. [PMID: 35085699 DOI: 10.1016/j.jconrel.2022.01.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/23/2021] [Accepted: 01/19/2022] [Indexed: 12/25/2022]
Abstract
Human insulin (HI) has fascinating metal-facilitated self-assembly properties that are essential for its biological function. HI has a natural Zn2+ binding site and we have previously shown that covalently attached abiotic ligands (e.g., bipyridine, terpyridine) can lead to the formation of nanosized oligomeric structures through the coordination of metal ions. Here we studied the hypothesis that metal ions can be used to directly control the pharmacokinetics of insulin after covalent attachment of an abiotic ligand that binds metal ions. We evaluated the pharmacokinetics (PK) and biodistribution of HI self-assemblies directed by metal ion coordination (i.e., Fe2+/Zn2+, Eu3+/Zn2+, Fe2+/Co3+) using preclinical SPECT/CT imaging and ex vivo gamma counting. HI was site-specifically modified with terpyridine (Tpy) at the PheB1 or LysB29 position to create conjugates that bind either Fe2+ or Eu3+, while its natural binding site (e.g., HisB10) preferentially coordinates with either Zn2+ or Co3+. HI was also functionalized with trans-cyclooctene (TCO) opposite to Tpy at PheB1 or LysB29, respectively, to allow for tetrazine-TCO coupling via a tetrazine-modified DTPA followed by 111In-radiolabeling for SPECT/CT imaging. When the 111In-B29Tpy-HI conjugate was coordinated with Fe2+/Zn2+, its retention at the injection site 6 h after injection was ~8-fold higher than the control without the metal ions, while its kidney accumulation was lower. 111In-B1Tpy-HI showed comparable retention at the injection site 6 h after injection and slightly increased retention at 24 h. However, higher kidney accumulation and residence time of degraded 111In-B1Tpy-HI was observed compared to that of 111In-B29Tpy-HI. Quantitative PK analysis based on SPECT/CT images confirmed slower distribution from the injection site of the HI-metal ion assemblies compared to control HI conjugates. Our results show that the Tpy-binding site (i.e., PheB1 or LysB29) on HI and its coordination with the added metal ions (i.e., Fe2+/Zn2+ or Fe2+/Co3+) directed the distribution half-life of HI significantly. This clearly indicates that the PK of insulin can be controlled by complexation with different metal ions.
Collapse
Affiliation(s)
- Gokce Engudar
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Cristina Rodríguez-Rodríguez
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada; Department of Physics and Astronomy, Faculty of Science, University of British Columbia, 6224 Agricultural Road, Vancouver, BC V6T 1Z1, Canada
| | - Narendra Kumar Mishra
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark
| | - Marta Bergamo
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Guillaume Amouroux
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Knud J Jensen
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark.
| | - Katayoun Saatchi
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada.
| | - Urs O Häfeli
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada; Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| |
Collapse
|
43
|
Zhang Y, Lin Q, Wang T, Shi D, Fu Z, Si Z, Xu Z, Cheng Y, Shi H, Cheng D. Targeting Infiltrating Myeloid Cells in Gastric Cancer Using a Pretargeted Imaging Strategy Based on Bio-Orthogonal Diels-Alder Click Chemistry and Comparison with 89Zr-Labeled Anti-CD11b Positron Emission Tomography Imaging. Mol Pharm 2022; 19:246-257. [PMID: 34816721 DOI: 10.1021/acs.molpharmaceut.1c00745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Gastric cancer (GC) is a common cancer worldwide, with high incidence and mortality rates. Therefore, early and precise diagnosis is critical to improving GC prognosis. Tumor-associated myeloid cells infiltrate the tumor microenvironment (TME) and can produce immunosuppressive effects in the early stage of the tumor. The surface integrin receptor CD11b is widely expressed in the specific subsets of myeloid cells, and it has the characteristics of high abundance, high specificity, and high potential for targeted immunotherapy. In this study, two strategies for labeling anti-CD11b, including 89Zr-DFO-anti-CD11b and pretargeted imaging (68Ga-NOTA-polypeptide-PEG11-Tz/anti-CD11b-TCO), were used to evaluate the value of early diagnosis of GC and confirm the advantages of the pretargeted strategy for the diagnosis of GC. Pretargeted molecular probe 68Ga-NOTA-polypeptide-PEG11-Tz was synthesized. The binding affinity of the Tz-radioligand to CD11b was evaluated in vitro, and its blood pharmacokinetic test was performed in vivo. Moreover, the anti-CD11b antibody was conjugated with a p-isothiocyanatobenzyl-desferrioxamine (SCN-DFO) chelator and radiolabeled with zirconium-89. Biodistribution and positron-emission computed tomography imaging experiments were performed in MGC-803 tumor-bearing model mice to evaluate the value of the early diagnosis of GC. Histological evaluation of MGC-803 tumors was conducted to confirm the infiltration of the GC TME with CD11b+ myeloid cells. 68Ga-NOTA-polypeptide-PEG11-Tz was successfully radiosynthesized, with the radiochemical purity above 95%, as confirmed by reversed-phase high-performance liquid chromatography. The radioligand showed favorable stability in normal saline and phosphate-buffered saline, good affinity to RAW264.7 cells, and rapid blood clearance in mice. The results of biodistribution and imaging experiments using the pretargeted method showed that the tumor/muscle ratios were 5.17 ± 2.98, 5.94 ± 1.46, and 4.46 ± 2.73 at the pretargeting intervals of 24, 48, and 72 h, respectively. The experimental results using the method of the directly labeling antibody (89Zr-DFO-anti-CD11b) showed that, despite radioactive accumulation in the tumor, there was a higher level of radioactive accumulation in normal tissues. The tumor/muscle ratios were 1.09 ± 0.67, 1.66 ± 0.95, 2.94 ± 1.24, 3.64 ± 1.21, and 3.55 ± 1.64 at 1, 24, 48, 72, and 120 h. The current research proved the value of 68Ga-NOTA-polypeptide-PEG11-Tz/anti-CD11b-TCO in the diagnosis of GC using the pretargeted strategy. Compared to 89Zr-DFO-anti-CD11b, the image contrast achieved by the pretargeted strategy was relatively improved, and the background accumulation of the probe was relatively low. These advantages can improve the diagnostic efficiency for GC and provide supporting evidence for radioimmunotherapy targeting CD11b receptors.
Collapse
Affiliation(s)
- Yingying Zhang
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032, People's Republic of China
| | - Qingyu Lin
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032, People's Republic of China.,Institute of Nuclear Medicine, Fudan University, Shanghai 200032, People's Republic of China.,Shanghai Institute of Medical Imaging, Shanghai 200032, People's Republic of China
| | - Tingting Wang
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032, People's Republic of China
| | - Dai Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032, People's Republic of China
| | - Zhequan Fu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032, People's Republic of China
| | - Zhan Si
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032, People's Republic of China
| | - Zhan Xu
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032, People's Republic of China
| | - Yuan Cheng
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032, People's Republic of China
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032, People's Republic of China.,Institute of Nuclear Medicine, Fudan University, Shanghai 200032, People's Republic of China.,Shanghai Institute of Medical Imaging, Shanghai 200032, People's Republic of China
| | - Dengfeng Cheng
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai 200032, People's Republic of China.,Institute of Nuclear Medicine, Fudan University, Shanghai 200032, People's Republic of China.,Shanghai Institute of Medical Imaging, Shanghai 200032, People's Republic of China
| |
Collapse
|
44
|
Sun H, Xue Q, Zhang C, Wu H, Feng P. Derivatization based on tetrazine scaffolds: synthesis of tetrazine derivatives and their biomedical applications. Org Chem Front 2022. [DOI: 10.1039/d1qo01324f] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The recent advances in tetrazine scaffold-based derivatizations have been summarized. The advantages and limitations of derivatization methods and applications of the developed tetrazine derivatives in bioorthogonal chemistry have been highlighted.
Collapse
Affiliation(s)
- Hongbao Sun
- Huaxi MR Research Center (HMRRC), Department of Radiology, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qinghe Xue
- Huaxi MR Research Center (HMRRC), Department of Radiology, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chang Zhang
- Huaxi MR Research Center (HMRRC), Department of Radiology, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Haoxing Wu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ping Feng
- Clinical Trial Center, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
45
|
Shalgunov V, Engudar G, Bohrmann L, Wharton L, Maskell K, Johann K, Barz M, Schaffer P, Herth MM, Radchenko V. Radiolabeling of a polypeptide polymer for intratumoral delivery of alpha-particle emitter, 225Ac, and beta-particle emitter, 177Lu. Nucl Med Biol 2021; 104-105:11-21. [PMID: 34839209 DOI: 10.1016/j.nucmedbio.2021.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/22/2021] [Accepted: 11/08/2021] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Radiotherapy of cancer requires both alpha- and beta-particle emitting radionuclides, as these radionuclide types are efficient at destroying different types of tumors. Both classes of radionuclides require a vehicle, such as an antibody or a polymer, to be delivered and retained within the tumor. Polyglutamic acid (pGlu) is a polymer that has proven itself effective as a basis of drug-polymer conjugates in the clinic, while its derivatives have been used for pretargeted tumor imaging in a research setup. trans-Cyclooctene (TCO) modified pGlu is suitable for pretargeted imaging or therapy, as well as for intratumoral radionuclide therapy. In all cases, it becomes indirectly radiolabeled via the bioorthogonal click reaction with the tetrazine (Tz) molecule carrying the radionuclide. In this study, we report the radiolabeling of TCO-modified pGlu with either lutetium-177 (177Lu), a beta-particle emitter, or actinium-225 (225Ac), an alpha-particle emitter, using the click reaction between TCO and Tz. METHODS A panel of Tz derivatives containing a metal ion binding chelator (DOTA or macropa) connected to the Tz moiety directly or through a polyethylene glycol (PEG) linker was synthesized and tested for their ability to chelate 177Lu and 225Ac, and click to pGlu-TCO. Radiolabeled 177Lu-pGlu and 225Ac-pGlu were isolated by size exclusion chromatography. The retention of 177Lu or 225Ac by the obtained conjugates was investigated in vitro in human serum. RESULTS All DOTA-modified Tzs efficiently chelated 177Lu resulting in average radiochemical conversions (RCC) of >75%. Isolated radiochemical yields (RCY) for 177Lu-pGlu prepared from 177Lu-Tzs ranged from 31% to 55%. TLC analyses detected <5% unchelated 177Lu for all 177Lu-pGlu preparations over six days in human serum. For 225Ac chelation, optimized RCCs ranged from 61 ± 34% to quantitative for DOTA-Tzs and were quantitative for the macropa-modified Tz (>98%). Isolated radiochemical yields (RCY) for 225Ac-pGlu prepared from 225Ac-Tzs ranged from 28% to 51%. For 3 out of 5 225Ac-pGlu conjugates prepared from DOTA-Tzs, the amount of unchelated 225Ac stayed below 10% over six days in human serum, while 225Ac-pGlu prepared from macropa-Tz showed a steady release of up to 37% 225Ac. CONCLUSION We labeled TCO-modified pGlu polymers with alpha- and beta-emitting radionuclides in acceptable RCYs. All 177Lu-pGlu preparations and some 225Ac-pGlu preparations showed excellent stability in human plasma. Our work shows the potential of pGlu as a vehicle for alpha- and beta-radiotherapy of tumors and demonstrated the usefulness of Tz ligation for indirect radiolabeling.
Collapse
Affiliation(s)
- Vladimir Shalgunov
- Department for Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Gokce Engudar
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
| | - Lennart Bohrmann
- Department for Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Luke Wharton
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada; Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z, Canada
| | - Keiran Maskell
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada; Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 0A7, Canada
| | - Kerstin Johann
- Department of Chemistry, Johannes Gutenberg University, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Matthias Barz
- Department of Chemistry, Johannes Gutenberg University, Duesbergweg 10-14, 55128 Mainz, Germany; Division of Biotherapeutics, Leiden Academic Center for Drug Research (LACDR), Einsteinweg 55, 2333CC Leiden, the Netherlands
| | - Paul Schaffer
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada; Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 0A7, Canada; Department of Radiology, University of British Columbia, 2775 Lauret St, Vancouver, BC V5Z 1M9, Canada
| | - Matthias M Herth
- Department for 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.
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada; Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z, Canada.
| |
Collapse
|
46
|
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
|
47
|
Sun L, Gai Y, Li Z, Zhang X, Li J, Ma Y, Li H, Barajas RJ, Zeng D. Development of Dual Receptor Enhanced Pre-Targeting Strategy-A Novel Promising Technology for Immuno-Positron Emission Tomography Imaging. ADVANCED THERAPEUTICS 2021; 4:2100110. [PMID: 35309962 PMCID: PMC8932640 DOI: 10.1002/adtp.202100110] [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: 05/17/2021] [Indexed: 11/06/2022]
Abstract
PET imaging has become an important diagnostic tool in the era of precise medicine. Various pre-targeting systems have been reported to address limitations associated with traditional immuno-PET. However, the application of these mono-receptor based pre-targeting (MRPT) strategies is limited to non-internalizable antibodies, and the tumor uptake is usually much lower than that in the corresponding immuno-PET. To circumvent these limitations, we develop the first Dual-Receptor Pre-Targeting (DRPT) system through entrapping the tumor-receptor-specific radioligand by the pre-administered antibody. Besides the similar ligation pathway happens in MRPT, incorporation of a tumor-receptor-specific peptide into the radioligand in DRPT enhances both concentration and retention of the radioligand on tumor, promoting its ligation with pre-administered mAb on cell-surface and/or internalized into tumor-cells. In this study, 64Cu based DRPT shows superior performance over corresponding MRPT and immuno-PET using internalizable antibodies. Besides, the compatibility of DRPT with short-lived and generator-produced 68Ga is demonstrated, leveraging its advantage in reducing radio-dose exposure. Furthermore, the feasibility of reducing the amount of the pre-administered antibody is confirmed, indicating the cost saving potential of DRPT. In summary, synergizing advantages of dual-receptor targeting and pre-targeting, we expect that this DRPT strategy can become a breakthrough technology in the field of antibody-based molecular imaging.
Collapse
Affiliation(s)
- Lingyi Sun
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA; Center of Radiochemistry Research, Knight Cardiovascular Institute, Oregon Health & Science University, Portland 97239, USA
| | - Yongkang Gai
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA
| | - Zhonghan Li
- Center of Radiochemistry Research, Knight Cardiovascular Institute, Oregon Health & Science University, Portland 97239, USA
| | - Xiaohui Zhang
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA
| | - Jianchun Li
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA
| | - Yongyong Ma
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA
| | - Huiqiang Li
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA
| | - Ramon J Barajas
- Department of Diagnostic Radiology, Oregon Health & Science University, Portland 97239, USA; Advanced Imaging Research Center, Oregon Health & Science University, Portland 97239, USA; Translational Oncology Research Program, Knight Cancer Institute, Oregon Health & Science University, Portland 97239, USA
| | - Dexing Zeng
- Department of Radiology, University of Pittsburgh, Pittsburgh 15213, USA; Center of Radiochemistry Research, Knight Cardiovascular Institute, Oregon Health & Science University, Portland 97239, USA; Department of Diagnostic Radiology, Oregon Health & Science University, Portland 97239, USA
| |
Collapse
|
48
|
Bidesi NSR, Vang Andersen I, Windhorst AD, Shalgunov V, Herth MM. The role of neuroimaging in Parkinson's disease. J Neurochem 2021; 159:660-689. [PMID: 34532856 PMCID: PMC9291628 DOI: 10.1111/jnc.15516] [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: 05/12/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 11/29/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that affects millions of people worldwide. Two hallmarks of PD are the accumulation of alpha-synuclein and the loss of dopaminergic neurons in the brain. There is no cure for PD, and all existing treatments focus on alleviating the symptoms. PD diagnosis is also based on the symptoms, such as abnormalities of movement, mood, and cognition observed in the patients. Molecular imaging methods such as magnetic resonance imaging (MRI), single-photon emission computed tomography (SPECT), and positron emission tomography (PET) can detect objective alterations in the neurochemical machinery of the brain and help diagnose and study neurodegenerative diseases. This review addresses the application of functional MRI, PET, and SPECT in PD patients. We provide an overview of the imaging targets, discuss the rationale behind target selection, the agents (tracers) with which the imaging can be performed, and the main findings regarding each target's state in PD. Molecular imaging has proven itself effective in supporting clinical diagnosis of PD and has helped reveal that PD is a heterogeneous disorder, which has important implications for the development of future therapies. However, the application of molecular imaging for early diagnosis of PD or for differentiation between PD and atypical parkinsonisms has remained challenging. The final section of the review is dedicated to new imaging targets with which one can detect the PD-related pathological changes upstream from dopaminergic degeneration. The foremost of those targets is alpha-synuclein. We discuss the progress of tracer development achieved so far and challenges on the path toward alpha-synuclein imaging in humans.
Collapse
Affiliation(s)
- Natasha S R Bidesi
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Ida Vang Andersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Albert D Windhorst
- Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Matthias M Herth
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
49
|
Hu Y, Zhang J, Miao Y, Wen X, Wang J, Sun Y, Chen Y, Lin J, Qiu L, Guo K, Chen HY, Ye D. Enzyme-Mediated In Situ Self-Assembly Promotes In Vivo Bioorthogonal Reaction for Pretargeted Multimodality Imaging. Angew Chem Int Ed Engl 2021; 60:18082-18093. [PMID: 34010512 DOI: 10.1002/anie.202103307] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 05/01/2021] [Indexed: 12/13/2022]
Abstract
Pretargeted imaging has emerged as a promising approach to advance nuclear imaging of malignant tumors. Herein, we combine the enzyme-mediated fluorogenic reaction and in situ self-assembly with the inverse electron demand Diels-Alder (IEDDA) reaction to develop an activatable pretargeted strategy for multimodality imaging. The trans-cyclooctene (TCO) bearing small-molecule probe, P-FFGd-TCO, can be activated by alkaline phosphatase and in situ self-assembles into nanoaggregates (FMNPs-TCO) retained on the membranes, permitting to (1) amplify near-infrared (NIR) fluorescence (FL) and magnetic resonance imaging (MRI) signals, and (2) enrich TCOs to promote IEDDA ligation. The Gallium-68 (68 Ga) labeled tetrazine can readily conjugate the tumor-retained FMNPs-TCO to enhance radioactivity uptake in tumors. Strong NIR FL, MRI, and positron emission tomography (PET) signals are concomitantly achieved, allowing for pretargeted multimodality imaging of ALP activity in HeLa tumor-bearing mice.
Collapse
Affiliation(s)
- Yuxuan Hu
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Junya Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yinxing Miao
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, China
| | - Xidan Wen
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Jian Wang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, 211800, China
| | - Yidan Sun
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yinfei Chen
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, China
| | - Jianguo Lin
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, China
| | - Ling Qiu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, China
| | - Kai Guo
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, 211800, China
| | - Hong-Yuan Chen
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Deju Ye
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
50
|
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
|