1
|
Altai M, Nagy Á, Granit P, Zedan W, Cerezo-Magaña M, Park J, Lückerath K, Geres S, Sydoff M, Thorek DLJ, Westerlund K, Ulmert D, Karlström AE. Optimizing peptide nucleic acid-based pretargeting for enhanced targeted radionuclide therapy. J Control Release 2025; 381:113551. [PMID: 39986477 DOI: 10.1016/j.jconrel.2025.02.047] [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: 08/27/2024] [Revised: 01/17/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Radiolabeled targeting agents have emerged as valuable tools for the treatment of disseminated cancer. Monoclonal antibodies (mAbs) are widely employed as carriers for diagnostic and therapeutic radionuclides due to their exceptional specificity and affinity. However, their prolonged circulatory half-life can diminish diagnostic efficacy and increase radiation exposure to non-target tissues in therapeutic applications, resulting in dose-limiting toxicities. To overcome this limitation, pretargeting technologies emerge as promising strategies to enhance tumor-to-background ratio and reduce radiation exposure of healthy tissues. Our previous work introduced a pretargeting concept leveraging the specific interaction between two peptide nucleic acid (PNA) probes, HP1 and HP2, as the recognition mechanism. This early iteration of the PNA-based concept showed limited efficacy when used with mAb-based vectors. To improve its performance, we re-engineered the primary and secondary targeting agents by incorporating newly designed PNA-probes. As the primary targeting agent, we functionalized trastuzumab (T), a well-characterized human epidermal growth factor receptor 2 (HER2)-targeting IgG1 mAb, with a 9-mer PNA probe (HP9). Both FcIII-based covalent UV-light crosslinking and enzyme-mediated glyco-engineering click-chemistry methods were applied to generate trastuzumab-PNA conjugates T-FcIII-HP9 and T-gly-HP9, respectively. As a radionuclide-carrying secondary agent, we utilized a 9-mer complementary PNA probe, HP16, which forms a stable duplex with HP9 as well as displaying favorable in vivo kinetics. Biacore and flow cytometry assessment of the HP9-conjugated trastuzumab agents demonstrated retained HER2-binding properties. The secondary HP16 probe, labeled with either a dye or a radionuclide, showed cell surface accumulation contingent on the presence of HP9 on the primary HER2-targeting agents. In vivo, T-gly-HP9 exhibited significantly longer blood circulation half-life and superior tumor uptake compared to T-FcIII-HP9. Further, therapeutic dosing with [177Lu]-HP16 of trastuzumab-HP9 pretargeted HER2+ tumor models resulted in significantly delayed disease progression and extended survival compared to untreated subjects. Furthermore, pretargeted [177Lu]-HP16 exhibited comparable efficacy to [177Lu]-trastuzumab in both delaying disease progression and prolonging survival. In conclusion, the optimization of our PNA-based pretargeting system has resulted in exceptional in vivo targeting characteristics and therapeutic efficacy, validating the potential of this novel approach.
Collapse
Affiliation(s)
- Mohamed Altai
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden; Lund University Cancer Centre (LUCC), Lund University, Lund, Sweden
| | - Ábel Nagy
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Center, Stockholm, Sweden
| | - Pauline Granit
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Center, Stockholm, Sweden
| | - Wahed Zedan
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden; Lund University Cancer Centre (LUCC), Lund University, Lund, Sweden
| | - Myriam Cerezo-Magaña
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden; Lund University Cancer Centre (LUCC), Lund University, Lund, Sweden
| | - Julie Park
- Department of Molecular & Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America
| | - Katharina Lückerath
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, DKTK, Essen, Germany
| | - Susanne Geres
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Marie Sydoff
- Lund University Bioimaging Centre (LBIC), Lund University, Lund, Sweden
| | - Daniel L J Thorek
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States of America; Department of Biomedical Engineering, Washington University, St. Louis, MO, United States of America; Oncologic Imaging Program, Siteman Cancer Center, St. Louis, MO, United States of America
| | - Kristina Westerlund
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Center, Stockholm, Sweden
| | - David Ulmert
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden; Lund University Cancer Centre (LUCC), Lund University, Lund, Sweden; Department of Molecular & Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America
| | - Amelie Eriksson Karlström
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Center, Stockholm, Sweden.
| |
Collapse
|
2
|
Fayn S, Roy S, Cabalteja CC, Lee W, Makala H, Baidoo K, Nambiar D, Sheehan‐Klenk J, Chung J, Buffington J, Ho M, Escorcia FE, Cheloha RW. Generation of Site-Specifically Labeled Affinity Reagents via Use of a Self-Labeling Single Domain Antibody. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417160. [PMID: 39965119 PMCID: PMC11984916 DOI: 10.1002/advs.202417160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/30/2025] [Indexed: 02/20/2025]
Abstract
Several chemical and enzymatic methods have been used to link antibodies to moieties that facilitate visualization of cognate targets. Emerging evidence suggests that the extent of labeling, dictated by the type of chemistry used, has a substantial impact on performance, especially in the context of antibodies used for the visualization of tumors in vivo. These effects are particularly pronounced in studies using small antibody fragments, such as single-domain antibodies, or nanobodies. Here, we leverage a new variety of conjugation chemistry, based on a nanobody that forms a crosslink with a specialized high-affinity epitope analogue, to label target-specific nanobody constructs with functionalities of choice, including fluorophores, chelators, and click chemistry handles. Using heterodimeric nanobody conjugates, comprised of an antigen recognition module and a self-labeling module, enables us to attach the desired functional group at a location distal to the site of antigen recognition. Constructs generated using this approach bound to antigens expressed on xenograft murine models of liver cancer and allowed for non-invasive diagnostic imaging. The modularity of our approach using a self-labeling nanobody offers a novel method for site-specific functionalization of biomolecules and can be extended to other applications for which covalent labeling is required.
Collapse
Affiliation(s)
- Stanley Fayn
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
- Oxford Institute for Radiation OncologyDepartment of OncologyUniversity of OxfordOxfordOX3 7DQUK
| | - Swarnali Roy
- Chemical Biology in Signaling SectionNational Institutes of Diabetes and Digestive and Kidney DiseasesNational Institutes of HealthBethesdaMD20892USA
| | - Chino C. Cabalteja
- Chemical Biology in Signaling SectionNational Institutes of Diabetes and Digestive and Kidney DiseasesNational Institutes of HealthBethesdaMD20892USA
| | - Woonghee Lee
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Hima Makala
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Kwamena Baidoo
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Divya Nambiar
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Julia Sheehan‐Klenk
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Joon‐Yong Chung
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Jesse Buffington
- Antibody Engineering ProgramCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Mitchell Ho
- Antibody Engineering ProgramCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
- Laboratory of Molecular BiologyCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Freddy E. Escorcia
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
- Radiation Oncology BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Ross W. Cheloha
- Chemical Biology in Signaling SectionNational Institutes of Diabetes and Digestive and Kidney DiseasesNational Institutes of HealthBethesdaMD20892USA
| |
Collapse
|
3
|
Saccullo E, Patamia V, Tomarchio EG, Zagni C, Floresta G, Rescifina A. Unveiling the chemistry of antibody conjugation for enhanced PET imaging: Current trends and future directions. Bioorg Chem 2025; 155:108115. [PMID: 39756200 DOI: 10.1016/j.bioorg.2024.108115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/23/2024] [Accepted: 12/28/2024] [Indexed: 01/07/2025]
Abstract
Positron Emission Tomography (PET) has emerged as a powerful imaging technique in molecular medicine, enabling the non-invasive visualisation and quantification of biological processes at the molecular level. Antibody-based PET imaging has recently gained prominence, offering specific targeting capabilities for various diseases. This scientific article delves into the intricate chemistry underlying antibody conjugation strategies for PET, providing a comprehensive understanding of the key principles and advancements in this rapidly evolving field. The article begins with a detailed exploration of various antibody conjugation methodologies, encompassing both covalent and non-covalent approaches. The chemical intricacies of bioconjugation reactions, such as amine and thiol chemistry, click chemistry, and bioorthogonal chemistry, are thoroughly discussed in the context of antibody modification. Additionally, the article critically analyses recent advancements in radiolabeling strategies for PET, including using radionuclides with favourable decay characteristics. This discussion covers both traditional radioisotopes and emerging alternatives, demonstrating their potential to raise the effectiveness of PET imaging agents based on antibodies. Ultimately, this article aims to contribute to the ongoing efforts to advance the field toward more effective diagnostic tools for personalized medicine.
Collapse
Affiliation(s)
- Erika Saccullo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy; Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy
| | - Vincenzo Patamia
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy.
| | - Elisabetta Grazia Tomarchio
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy; Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy
| | - Chiara Zagni
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy
| | - Giuseppe Floresta
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy.
| | - Antonio Rescifina
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
4
|
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
|
5
|
Delaney S, Nagy Á, Karlström AE, Zeglis BM. Site-Specific Photoaffinity Bioconjugation for the Creation of 89Zr-Labeled Radioimmunoconjugates. Mol Imaging Biol 2023; 25:1104-1114. [PMID: 37052759 PMCID: PMC10570397 DOI: 10.1007/s11307-023-01818-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/14/2023] [Accepted: 03/23/2023] [Indexed: 04/14/2023]
Abstract
PURPOSE Site-specific approaches to bioconjugation produce well-defined and homogeneous immunoconjugates with potential for superior in vivo behavior compared to analogs synthesized using traditional, stochastic methods. The possibility of incorporating photoaffinity chemistry into a site-specific bioconjugation strategy is particularly enticing, as it could simplify and accelerate the preparation of homogeneous immunoconjugates for the clinic. In this investigation, we report the synthesis, in vitro characterization, and in vivo evaluation of a site-specifically modified, 89Zr-labeled radioimmunoconjugate created via the reaction between an mAb and an Fc-binding protein bearing a photoactivatable 4-benzoylphenylalanine residue. PROCEDURES A variant of the Fc-binding Z domain of protein A containing a photoactivatable, 4-benzoylphenylalanine residue - Z(35BPA) - was modified with desferrioxamine (DFO), combined with the A33 antigen-targeting mAb huA33, and irradiated with UV light. The resulting immunoconjugate - DFOZ(35BPA)-huA33 - was purified and characterized via SDS-PAGE, MALDI-ToF mass spectrometry, surface plasmon resonance, and flow cytometry. The radiolabeling of DFOZ(35BPA)-huA33 was optimized to produce [89Zr]Zr-DFOZ(35BPA)-huA33, and the immunoreactivity of the radioimmunoconjugate was determined with SW1222 human colorectal cancer cells. Finally, the in vivo performance of [89Zr]Zr-DFOZ(35BPA)-huA33 in mice bearing subcutaneous SW1222 xenografts was interrogated via PET imaging and biodistribution experiments and compared to that of a stochastically labeled control radioimmunoconjugate, [89Zr]Zr-DFO-huA33. RESULTS HuA33 was site-specifically modified with Z(35BPA)-DFO, producing an immunoconjugate with on average 1 DFO/mAb, high in vitro stability, and high affinity for its target. [89Zr]Zr-DFOZ(35BPA)-huA33 was synthesized in 95% radiochemical yield and exhibited a specific activity of 2 mCi/mg and an immunoreactive fraction of ~ 0.85. PET imaging and biodistribution experiments revealed that high concentrations of the radioimmunoconjugate accumulated in tumor tissue (i.e., ~ 40%ID/g at 120 h p.i.) but also that the Z(35BPA)-bearing immunoPET probe produced higher uptake in the liver, spleen, and kidneys than its stochastically modified cousin, [89Zr]Zr-DFO-huA33. CONCLUSIONS Photoaffinity chemistry and an Fc-binding variant of the Z domain were successfully leveraged to create a novel site-specific strategy for the synthesis of radioimmunoconjugates. The probe synthesized using this method - DFOZ(35BPA)-huA33 - was well-defined and homogeneous, and the resulting radioimmunoconjugate ([89Zr]Zr-DFOZ(35BPA)-huA33) boasted high specific activity, stability, and immunoreactivity. While the site-specifically modified radioimmunoconjugate produced high activity concentrations in tumor tissue, it also yielded higher uptake in healthy organs than a stochastically modified analog, suggesting that optimization of this system is necessary prior to clinical translation.
Collapse
Affiliation(s)
- Samantha Delaney
- Department of Chemistry, Hunter College, City University of New York, New York, NY, USA
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ábel Nagy
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Amelie Eriksson Karlström
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden.
| | - Brian M Zeglis
- Department of Chemistry, Hunter College, City University of New York, New York, NY, USA.
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY, USA.
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY, USA.
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
6
|
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
|
7
|
Bauer D, Sarrett SM, Lewis JS, Zeglis BM. Click chemistry: a transformative technology in nuclear medicine. Nat Protoc 2023; 18:1659-1668. [PMID: 37100960 PMCID: PMC10293801 DOI: 10.1038/s41596-023-00825-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/10/2023] [Indexed: 04/28/2023]
Abstract
The 2022 Nobel Prize in Chemistry was awarded to Professors K. Barry Sharpless, Morten Meldal and Carolyn Bertozzi for their pioneering roles in the advent of click chemistry. Sharpless and Meldal worked to develop the canonical click reaction-the copper-catalyzed azide-alkyne cycloaddition-while Bertozzi opened new frontiers with the creation of the bioorthogonal strain-promoted azide-alkyne cycloaddition. These two reactions have revolutionized chemical and biological science by facilitating selective, high yielding, rapid and clean ligations and by providing unprecedented ways to manipulate living systems. Click chemistry has affected every aspect of chemistry and chemical biology, but few disciplines have been impacted as much as radiopharmaceutical chemistry. The importance of speed and selectivity in radiochemistry make it an almost tailor-made application of click chemistry. In this Perspective, we discuss the ways in which the copper-catalyzed azide-alkyne cycloaddition, the strain-promoted azide-alkyne cycloaddition and a handful of 'next-generation' click reactions have transformed radiopharmaceutical chemistry, both as tools for more efficient radiosyntheses and as linchpins of technologies that have the potential to improve nuclear medicine.
Collapse
Affiliation(s)
- David Bauer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samantha M Sarrett
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Chemistry, Hunter College of the City University of New York, New York, NY, USA
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY, USA
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA.
| | - Brian M Zeglis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Chemistry, Hunter College of the City University of New York, New York, NY, USA.
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY, USA.
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA.
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY, USA.
| |
Collapse
|
8
|
Zhong X, Yan J, Ding X, Su C, Xu Y, Yang M. Recent Advances in Bioorthogonal Click Chemistry for Enhanced PET and SPECT Radiochemistry. Bioconjug Chem 2023; 34:457-476. [PMID: 36811499 DOI: 10.1021/acs.bioconjchem.2c00583] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Due to their high reaction rate and reliable selectivity, bioorthogonal click reactions have been extensively investigated in numerous research fields, such as nanotechnology, drug delivery, molecular imaging, and targeted therapy. Previous reviews on bioorthogonal click chemistry for radiochemistry mainly focus on 18F-labeling protocols employed to produce radiotracers and radiopharmaceuticals. In fact, besides fluorine-18, other radionuclides such as gallium-68, iodine-125, and technetium-99m are also used in the field of bioorthogonal click chemistry. Herein, to provide a more comprehensive perspective, we provide a summary of recent advances in radiotracers prepared using bioorthogonal click reactions, including small molecules, peptides, proteins, antibodies, and nucleic acids as well as nanoparticles based on these radionuclides. The combination of pretargeting with imaging modalities or nanoparticles, as well as the clinical translations study, are also discussed to illustrate the effects and potential of bioorthogonal click chemistry for radiopharmaceuticals.
Collapse
Affiliation(s)
- Xinlin Zhong
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P. R. China
- School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, P. R. China
| | - Junjie Yan
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Xiang Ding
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Chen Su
- Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi 214002, P. R. China
| | - Yuping Xu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Min Yang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P. R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
- School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, P. R. China
| |
Collapse
|
9
|
Melendez-Alafort L, Ferro-Flores G, De Nardo L, Ocampo-García B, Bolzati C. Zirconium immune-complexes for PET molecular imaging: Current status and prospects. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.215005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
10
|
MacPherson DS, Hwang D, Sarrett SM, Keinänen O, Rodriguez C, Rader C, Zeglis BM. Leveraging a Dual Variable Domain Immunoglobulin to Create a Site-Specifically Modified Radioimmunoconjugate. Mol Pharm 2023; 20:775-782. [PMID: 36377696 PMCID: PMC10263003 DOI: 10.1021/acs.molpharmaceut.2c00700] [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] [Indexed: 11/16/2022]
Abstract
Site-specifically modified radioimmunoconjugates exhibit superior in vitro and in vivo behavior compared to analogues synthesized via traditional stochastic methods. However, the development of approaches to site-specific bioconjugation that combine high levels of selectivity, simple reaction conditions, and clinical translatability remains a challenge. Herein, we describe a novel solution to this problem: the use of dual-variable domain immunoglobulins (DVD-IgG). More specifically, we report the synthesis, in vitro evaluation, and in vivo validation of a 177Lu-labeled radioimmunoconjugate based on HER2DVD, a DVD-IgG containing the HER2-targeting variable domains of trastuzumab and the catalytic variable domains of IgG h38C2. To this end, we first modified HER2DVD with a phenyloxadiazolyl methlysulfone-modified variant of the chelator CHX-A″-DTPA (PODS-CHX-A''-DTPA) and verified the site-specificity of the conjugation for the reactive lysines within the catalytic domains via chemical assay, MALDI-ToF mass spectrometry, and SDS-PAGE. The chelator-bearing immunoconjugate was subsequently labeled with [177Lu]Lu3+ to produce the completed radioimmunoconjugate, [177Lu]Lu-CHX-A″-DTPAPODS-HER2DVD, in >80% radiochemical conversion and a specific activity of 29.5 ± 7.1 GBq/μmol. [177Lu]Lu-CHX-A″-DTPAPODS-HER2DVD did not form aggregates upon prolonged incubation in human serum, displayed 87% stability to demetalation over a 7 days of incubation in serum, and exhibited an immunoreactive fraction of 0.95 with HER2-coated beads. Finally, we compared the pharmacokinetic profile of [177Lu]Lu-CHX-A″-DTPAPODS-HER2DVD to that of a 177Lu-labeled variant of trastuzumab in mice bearing subcutaneous HER2-expressing BT-474 human breast cancer xenografts. The in vivo performance of [177Lu]Lu-CHX-A″-DTPAPODS-HER2DVD matched that of 177Lu-labeled trastuzumab, with the former producing a tumoral activity concentration of 34.1 ± 12.1 %ID/g at 168 h and tumor-to-blood, tumor-to-liver, and tumor-to-kidney activity concentration ratios of 10.5, 9.6, and 21.8, respectively, at the same time point. Importantly, the DVD-IgG did not exhibit a substantially longer serum half-life than the traditional IgG despite its significantly larger size (202 kDa for the former vs 148 kDa for the latter). Taken together, these data suggest that DVD-IgGs represent a viable platform for the future development of highly effective site-specifically labeled radioimmunoconjugates for diagnostic imaging, theranostic imaging, and radioimmunotherapy.
Collapse
Affiliation(s)
- Douglas S. MacPherson
- Department of Chemistry, Hunter College of the City University of New York, 413 East 69th Street, New York, New York 10028, United States
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, New York 10016, United States
- Advanced Science Research Center (ASRC) at The Graduate Center, City University of New York, 85 St. Nicholas Terrace, New York, NY 10031, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
| | - Dobeen Hwang
- Department of Immunology and Microbiology, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458, United States
| | - Samantha M. Sarrett
- Department of Chemistry, Hunter College of the City University of New York, 413 East 69th Street, New York, New York 10028, United States
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, New York 10016, United States
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
| | - Outi Keinänen
- Department of Chemistry, Hunter College of the City University of New York, 413 East 69th Street, New York, New York 10028, United States
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
- Department of Chemistry, University of Helsinki, P.O. Box 55, FI-00014 Helsinki, Finland
| | - Cindy Rodriguez
- Department of Chemistry, Hunter College of the City University of New York, 413 East 69th Street, New York, New York 10028, United States
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, New York 10016, United States
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
| | - Christoph Rader
- Department of Immunology and Microbiology, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458, United States
| | - Brian M. Zeglis
- Department of Chemistry, Hunter College of the City University of New York, 413 East 69th Street, New York, New York 10028, United States
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, New York 10016, United States
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, New York 10016, United States
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
- Department of Radiology, Weill Cornell Medical College, 520 East 70th Street, New York, New York 10065, United States
| |
Collapse
|
11
|
Lopes van den Broek S, García-Vázquez R, Andersen IV, Valenzuela-Nieto G, Shalgunov V, Battisti UM, Schwefel D, Modhiran N, Kramer V, Cheuquemilla Y, Jara R, Salinas-Varas C, Amarilla AA, Watterson D, Rojas-Fernandez A, Herth MM. Development and evaluation of an 18F-labeled nanobody to target SARS-CoV-2's spike protein. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2022; 2:1033697. [PMID: 39354971 PMCID: PMC11440877 DOI: 10.3389/fnume.2022.1033697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/21/2022] [Indexed: 10/03/2024]
Abstract
COVID-19, caused by the SARS-CoV-2 virus, has become a global pandemic that is still present after more than two years. COVID-19 is mainly known as a respiratory disease that can cause long-term consequences referred to as long COVID. Molecular imaging of SARS-CoV-2 in COVID-19 patients would be a powerful tool for studying the pathological mechanisms and viral load in different organs, providing insights into the disease and the origin of long-term consequences and assessing the effectiveness of potential COVID-19 treatments. Current diagnostic methods used in the clinic do not allow direct imaging of SARS-CoV-2. In this work, a nanobody (NB) - a small, engineered protein derived from alpacas - and an Fc-fused NB which selectively target the SARS-CoV-2 Spike protein were developed as imaging agents for positron emission tomography (PET). We used the tetrazine ligation to 18F-label the NB under mild conditions once the NBs were successfully modified with trans-cyclooctenes (TCOs). We confirmed binding to the Spike protein by SDS-PAGE. Dynamic PET scans in rats showed excretion through the liver for both constructs. Future work will evaluate in vivo binding to the Spike protein with our radioligands.
Collapse
Affiliation(s)
- Sara Lopes van den Broek
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rocío García-Vázquez
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ida Vang Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Guillermo Valenzuela-Nieto
- Institute of Medicine, Faculty of Medicine & Center for Interdisciplinary Studies on the Nervous System, CISNE, Universidad Austral de Chile, Valdivia, Chile
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Umberto M. Battisti
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - David Schwefel
- Institute of Medical Physics and Biophysics, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Naphak Modhiran
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia
| | | | - Yorka Cheuquemilla
- Institute of Medicine, Faculty of Medicine & Center for Interdisciplinary Studies on the Nervous System, CISNE, Universidad Austral de Chile, Valdivia, Chile
| | - Ronald Jara
- Institute of Medicine, Faculty of Medicine & Center for Interdisciplinary Studies on the Nervous System, CISNE, Universidad Austral de Chile, Valdivia, Chile
| | - Constanza Salinas-Varas
- Institute of Medicine, Faculty of Medicine & Center for Interdisciplinary Studies on the Nervous System, CISNE, Universidad Austral de Chile, Valdivia, Chile
| | - Alberto A. Amarilla
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia
| | - Daniel Watterson
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia
| | - Alejandro Rojas-Fernandez
- Institute of Medicine, Faculty of Medicine & Center for Interdisciplinary Studies on the Nervous System, CISNE, Universidad Austral de Chile, Valdivia, Chile
- Berking Biotechnology, Valdivia, Chile
| | - 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 & PET, Rigshospitalet Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
12
|
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
|
13
|
Zhao G, Li Z, Zhang R, Zhou L, Zhao H, Jiang H. Tetrazine bioorthogonal chemistry derived in vivo imaging. Front Mol Biosci 2022; 9:1055823. [PMID: 36465558 PMCID: PMC9709424 DOI: 10.3389/fmolb.2022.1055823] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 10/26/2022] [Indexed: 09/02/2023] Open
Abstract
Bioorthogonal chemistry represents plenty of highly efficient and biocompatible reactions that proceed selectively and rapidly in biological situations without unexpected side reactions towards miscellaneous endogenous functional groups. Arise from the strict demands of physiological reactions, bioorthogonal chemical reactions are natively selective transformations that are rarely found in biological environments. Bioorthogonal chemistry has long been applied to tracking and real-time imaging of biomolecules in their physiological environments. Thereinto, tetrazine bioorthogonal reactions are particularly important and have increasing applications in these fields owing to their unique properties of easily controlled fluorescence or radiation off-on mechanism, which greatly facilitate the tracking of real signals without been disturbed by background. In this mini review, tetrazine bioorthogonal chemistry for in vivo imaging applications will be attentively appraised to raise some guidelines for prior tetrazine bioorthogonal chemical studies.
Collapse
Affiliation(s)
- Gaoxiang Zhao
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Cancer Institute, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhutie Li
- China United Test and Evaluation (Qingdao) Co. Ltd., Qingdao, China
| | - Renshuai Zhang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Cancer Institute, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liman Zhou
- Key Laboratory of Chemistry and Engineering of Forest Products, State Ethnic Affairs Commission, Guangxi Key Laboratory of Chemistry and Engineering of Forest Products, Guangxi Collaborative Innovation Center for Chemistry and Engineering of Forest Products, School of Chemistry and Chemical Engineering, Guangxi Minzu University, Nanning, China
| | - Haibo Zhao
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Department of Sports Medicine, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongfei Jiang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Cancer Institute, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
14
|
van den Broek SL, Shalgunov V, Herth MM. Transport of nanomedicines across the blood-brain barrier: Challenges and opportunities for imaging and therapy. BIOMATERIALS ADVANCES 2022; 141:213125. [PMID: 36182833 DOI: 10.1016/j.bioadv.2022.213125] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 06/16/2023]
Abstract
The blood-brain barrier (BBB) is a protective and semipermeable border of endothelial cells that prevents toxins and foreign bodies to enter and damage the brain. Unfortunately, the BBB also hampers the development of pharmaceuticals targeting receptors, enzymes, or other proteins that lie beyond this barrier. Especially large molecules, such as monoclonal antibodies (mAbs) or nanoparticles, are prevented to enter the brain. The limited passage of these molecules partly explains why nanomedicines - targeting brain diseases - have not made it into the clinic to a great extent. As nanomedicines can target a wide range of targets including protein isoforms and oligomers or potentially deliver cytotoxic drugs safely to their targets, a pathway to smuggle nanomedicines into the brain would allow to treat brain diseases that are currently considered 'undruggable'. In this review, strategies to transport nanomedicines over the BBB will be discussed. Their challenges and opportunities will be highlighted with respect to their use for molecular imaging or therapies. Several strategies have been explored for this thus far. For example, carrier-mediated and receptor-mediated transcytosis (RMT), techniques to disrupt the BBB, nasal drug delivery or administering nanomedicines directly into the brain have been explored. RMT has been the most widely and successfully explored strategy. Recent work on the use of focused ultrasound based BBB opening has shown great promise. For example, successful delivery of mAbs into the brain has been achieved, even in a clinical setting. As nanomedicines bear the potential to treat incurable brain diseases, drug delivery technologies that can deliver nanomedicines into the brain will play an essential role for future treatment options.
Collapse
Affiliation(s)
- Sara Lopes van den Broek
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Matthias M Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| |
Collapse
|
15
|
Lopes van den Broek S, Shalgunov V, García Vázquez R, Beschorner N, Bidesi NSR, Nedergaard M, Knudsen GM, Sehlin D, Syvänen S, Herth MM. Pretargeted Imaging beyond the Blood-Brain Barrier-Utopia or Feasible? Pharmaceuticals (Basel) 2022; 15:1191. [PMID: 36297303 PMCID: PMC9612205 DOI: 10.3390/ph15101191] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
Pretargeting is a promising nuclear imaging technique that allows for the usage of antibodies (Abs) with enhanced imaging contrast and reduced patient radiation burden. It is based on bioorthogonal chemistry with the tetrazine ligation-a reaction between trans-cyclooctenes (TCOs) and tetrazines (Tzs)-currently being the most popular reaction due to its high selectivity and reactivity. As Abs can be designed to bind specifically to currently 'undruggable' targets such as protein isoforms or oligomers, which play a crucial role in neurodegenerative diseases, pretargeted imaging beyond the BBB is highly sought after, but has not been achieved yet. A challenge in this respect is that large molecules such as Abs show poor brain uptake. Uptake can be increased by receptor mediated transcytosis; however, it is largely unknown if the achieved brain concentrations are sufficient for pretargeted imaging. In this study, we investigated whether the required concentrations are feasible to reach. As a model Ab, we used the bispecific anti-amyloid beta (Aβ) anti-transferrin receptor (TfR) Ab 3D6scFv8D3 and conjugated it to a different amount of TCOs per Ab and tested different concentrations in vitro. With this model in hand, we estimated the minimum required TCO concentration to achieve a suitable contrast between the high and low binding regions. The estimation was carried out using pretargeted autoradiography on brain sections of an Alzheimer's disease mouse model. Biodistribution studies in wild-type (WT) mice were used to correlate how different TCO/Ab ratios alter the brain uptake. Pretargeted autoradiography showed that increasing the number of TCOs as well as increasing the TCO-Ab concentration increased the imaging contrast. A minimum brain concentration of TCOs for pretargeting purposes was determined to be 10.7 pmol/g in vitro. Biodistribution studies in WT mice showed a brain uptake of 1.1% ID/g using TCO-3D6scFv8D3 with 6.8 TCO/Ab. According to our estimations using the optimal parameters, pretargeted imaging beyond the BBB is not a utopia. Necessary brain TCO concentrations can be reached and are in the same order of magnitude as required to achieve sufficient contrast. This work gives a first estimate that pretargeted imaging is indeed possible with antibodies. This could allow the imaging of currently 'undruggable' targets and therefore be crucial to monitor (e.g., therapies for intractable neurodegenerative diseases).
Collapse
Affiliation(s)
- 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
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Rocío García Vázquez
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Natalie Beschorner
- Center for Translational Neuromedicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Natasha S. R. Bidesi
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Gitte M. Knudsen
- Neurobiology Research Unit, Rigshospitalet Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Dag Sehlin
- Rudbeck Laboratory, Department of Public Health and Caring Sciences, University of Uppsala, Dag Hammarskjölds väg 20, 75185 Uppsala, Sweden
| | - Stina Syvänen
- Rudbeck Laboratory, Department of Public Health and Caring Sciences, University of Uppsala, Dag Hammarskjölds väg 20, 75185 Uppsala, Sweden
| | - 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
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Jallinoja VIJ, Carney BD, Zhu M, Bhatt K, Yazaki PJ, Houghton JL. Cucurbituril-Ferrocene: Host-Guest Based Pretargeted Positron Emission Tomography in a Xenograft Model. Bioconjug Chem 2021; 32:1554-1558. [PMID: 34156824 PMCID: PMC9153067 DOI: 10.1021/acs.bioconjchem.1c00280] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pretargeted positron emission tomography is a macromolecule-driven nuclear medicine technique that involves targeting a preadministered antigen target-bound macromolecule with a radioligand in vivo, aiming to minimize the overall radiation dose. This study investigates the use of antibody based host-guest chemistry methodology for pretargeted positron emission tomography. We hypothesize that the novel pretargeting approach reported here overcomes the challenges the current pretargeting platforms have with the in vivo stability and modularity of the pretargeting components. A cucurbit[7]uril host molecule modified, anti-carcinoembryonic antigen antibody (M5A; CB7-M5A) and a 68Ga-radiolabeled ferrocene guest radioligand ([68Ga]Ga-NOTA-PEG3-NMe2-Fc) were studied as potential host-guest chemistry pretargeting agents for positron emission tomography in BxPC3 xenografted nude mice. The viability of the platform was studied via in vivo biodistribution and positron emission tomography. Tumor uptake of [68Ga]Ga-NOTA-PEG3-NMe2-Fc was significantly higher in mice which received CB7-M5A prior to the radioligand injection (pretargeted) (3.3 ± 0.7%ID/g) compared to mice which only received the radioligand (nonpretargeted) (0.2 ± 0.1%ID/g).
Collapse
Affiliation(s)
- Vilma IJ Jallinoja
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, USA
- Department of Radiology, Stony Brook University, Stony Brook, New York, 11774, USA
| | - Brandon D Carney
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, USA
- Department of Radiology, Stony Brook University, Stony Brook, New York, 11774, USA
| | - Meiying Zhu
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, USA
| | - Kavita Bhatt
- Department of Radiology, Stony Brook University, Stony Brook, New York, 11774, USA
| | - Paul J Yazaki
- Beckman Institute, City of Hope, Duarte, California 91010, USA
| | - Jacob L Houghton
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, USA
- Department of Radiology, Stony Brook University, Stony Brook, New York, 11774, USA
| |
Collapse
|
19
|
Cheewawisuttichai T, Brichacek M. Development of a multifunctional neoglycoside auxiliary for applications in glycomics research. Org Biomol Chem 2021; 19:6613-6617. [PMID: 34264248 DOI: 10.1039/d1ob00941a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A novel, multifunctional, tetrazine-containing neoglycoside auxiliary has been synthesized in three steps and 28% overall yield. The oxyamine was conjugated with unprotected carbohydrates under aqueous conditions (pH = 4.7), with DMF as a cosolvent, to provide neoglycosides in yields ranging between 51% and 68%. This auxiliary displayed broad advantages in the isolation and purification of complex carbohydrate mixtures, compatibility during extension by glycosyltransferases, and direct conjugation to chemical probes. Furthermore, the auxiliary can be removed in 96% yield under acidic conditions (0.25% TFA in H2O) that leave glycosidic linkages intact. Thereby, the tetrazine-containing neoglycoside auxiliary can serve to facilitate future glycomics investigations.
Collapse
|
20
|
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
|
21
|
Farleigh M, Pham TT, Yu Z, Kim J, Sunassee K, Firth G, Forte N, Chudasama V, Baker JR, Long NJ, Rivas C, Ma MT. New Bifunctional Chelators Incorporating Dibromomaleimide Groups for Radiolabeling of Antibodies with Positron Emission Tomography Imaging Radioisotopes. Bioconjug Chem 2021; 32:1214-1222. [PMID: 33724798 PMCID: PMC8299457 DOI: 10.1021/acs.bioconjchem.0c00710] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/20/2021] [Indexed: 01/23/2023]
Abstract
Positron Emission Tomography (PET) imaging with antibody-based contrast agents frequently uses the radioisotopes [64Cu]Cu2+ and [89Zr]Zr4+. The macrobicyclic chelator commonly known as sarcophagine (sar) is ideal for labeling receptor-targeted biomolecules with [64Cu]Cu2+. The siderophore chelator, desferrioxamine-B (dfo), has been widely used to incorporate [89Zr]Zr4+ into antibodies. Here, we describe new bifunctional chelators of sar and dfo: these chelators have been functionalized with dibromomaleimides (dbm), that enable site-specific and highly stable attachment of molecular cargoes to reduced, solvent-accessible, interstrand native disulfide groups. The new sar-dbm and dfo-dbm derivatives can be easily conjugated with the IgG antibody trastuzumab via reaction with reduced interstrand disulfide groups to give site-specifically modified dithiomaleamic acid (dtm) conjugates, sar-dtm-trastuzumab and dfo-dtm-trastuzumab, in which interstrand disulfides are rebridged covalently with a small molecule linker. Both sar- and dfo-dtm-trastuzumab conjugates have been radiolabeled with [64Cu]Cu2+ and [89Zr]Zr4+, respectively, in near quantitative radiochemical yield (>99%). Serum stability studies, in vivo PET imaging, and biodistribution analyses using these radiolabeled immunoconjugates demonstrate that both [64Cu]Cu-sar-dtm-trastuzumab and [89Zr]Zr-dfo-dtm-trastuzumab possess high stability in biological milieu. Dibromomaleimide technology can be easily applied to enable stable, site-specific attachment of radiolabeled chelators, such as sar and dfo, to native interstrand disulfide regions of antibodies, enabling tracking of antibodies with PET imaging.
Collapse
Affiliation(s)
- Matthew Farleigh
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Truc Thuy Pham
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Zilin Yu
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Jana Kim
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Kavitha Sunassee
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - George Firth
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Nafsika Forte
- Department
of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| | - Vijay Chudasama
- Department
of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| | - James R. Baker
- Department
of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| | - Nicholas J. Long
- Department
of Chemistry, Imperial College London, Molecular
Sciences Research Hub, London W12 0BZ, U.K.
| | - Charlotte Rivas
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Michelle T. Ma
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| |
Collapse
|
22
|
Bolzati C, Spolaore B. Enzymatic Methods for the Site-Specific Radiolabeling of Targeting Proteins. Molecules 2021; 26:3492. [PMID: 34201280 PMCID: PMC8229434 DOI: 10.3390/molecules26123492] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 12/19/2022] Open
Abstract
Site-specific conjugation of proteins is currently required to produce homogenous derivatives for medicine applications. Proteins derivatized at specific positions of the polypeptide chain can actually show higher stability, superior pharmacokinetics, and activity in vivo, as compared with conjugates modified at heterogeneous sites. Moreover, they can be better characterized regarding the composition of the derivatization sites as well as the conformational and activity properties. To this aim, several site-specific derivatization approaches have been developed. Among these, enzymes are powerful tools that efficiently allow the generation of homogenous protein-drug conjugates under physiological conditions, thus preserving their native structure and activity. This review will summarize the progress made over the last decade on the use of enzymatic-based methodologies for the production of site-specific labeled immunoconjugates of interest for nuclear medicine. Enzymes used in this field, including microbial transglutaminase, sortase, galactosyltransferase, and lipoic acid ligase, will be overviewed and their recent applications in the radiopharmaceutical field will be described. Since nuclear medicine can benefit greatly from the production of homogenous derivatives, we hope that this review will aid the use of enzymes for the development of better radio-conjugates for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Cristina Bolzati
- Institute of Condensed Matter Chemistry and Technologies for Energy ICMATE-CNR, Corso Stati Uniti, 4, I-35127 Padova, Italy
| | - Barbara Spolaore
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via Marzolo, 5, I-35131 Padova, Italy
- CRIBI Biotechnology Center, University of Padua, Viale G. Colombo, 3, I-35131 Padova, Italy
| |
Collapse
|
23
|
Allott L, Amgheib A, Barnes C, Braga M, Brickute D, Wang N, Fu R, Ghaem-Maghami S, Aboagye EO. Radiolabelling an 18F biologic via facile IEDDA "click" chemistry on the GE FASTLab™ platform. REACT CHEM ENG 2021; 6:1070-1078. [PMID: 34123410 PMCID: PMC8167423 DOI: 10.1039/d1re00117e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023]
Abstract
The use of biologics in positron emission tomography (PET) imaging is an important area of radiopharmaceutical development and new automated methods are required to facilitate their production. We report an automated radiosynthesis method to produce a radiolabelled biologic via facile inverse electron demand Diels-Alder (IEDDA) "click" chemistry on a single GE FASTLab™ cassette. We exemplified the method by producing a fluorine-18 radiolabelled interleukin-2 (IL2) radioconjugate from a trans-cyclooctene (TCO) modified IL2 precursor. The radioconjugate was produced using a fully automated radiosynthesis on a single FASTLab™ cassette in a decay-corrected radiochemical yield (RCY, d.c.) of 19.8 ± 2.6% in 110 min (from start of synthesis); the molar activity was 132.3 ± 14.6 GBq μmol-1. The in vitro uptake of [18F]TTCO-IL2 correlated with the differential receptor expression (CD25, CD122, CD132) in PC3, NK-92 and activated human PBMCs. The automated method may be adapted for the radiosynthesis of any TCO-modified protein via IEDDA chemistry.
Collapse
Affiliation(s)
- Louis Allott
- Comprehensive Cancer Imaging Centre, Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
- Positron Emission Tomography Research Centre, Faculty of Health Sciences, University of Hull Cottingham Road Kingston upon Hull HU6 7RX UK
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Hull Cottingham Road Kingston upon Hull HU6 7RX UK
| | - Ala Amgheib
- Comprehensive Cancer Imaging Centre, Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
- Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
| | - Chris Barnes
- Comprehensive Cancer Imaging Centre, Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
| | - Marta Braga
- Comprehensive Cancer Imaging Centre, Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
| | - Diana Brickute
- Comprehensive Cancer Imaging Centre, Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
| | - Ning Wang
- Comprehensive Cancer Imaging Centre, Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
| | - Ruisi Fu
- Comprehensive Cancer Imaging Centre, Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
| | - Sadaf Ghaem-Maghami
- Comprehensive Cancer Imaging Centre, Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
- Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
| | - Eric O Aboagye
- Comprehensive Cancer Imaging Centre, Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Du Cane Road London W12 0NN UK
| |
Collapse
|
24
|
Computational studies on the Carboni-Lindsey reaction. COMPUT THEOR CHEM 2021. [DOI: 10.1016/j.comptc.2021.113161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
25
|
Ferreira VFC, Oliveira BL, D'Onofrio A, Farinha CM, Gano L, Paulo A, Bernardes GJL, Mendes F. In Vivo Pretargeting Based on Cysteine-Selective Antibody Modification with IEDDA Bioorthogonal Handles for Click Chemistry. Bioconjug Chem 2020; 32:121-132. [PMID: 33295756 DOI: 10.1021/acs.bioconjchem.0c00551] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pretargeted imaging has emerged as an effective multistep strategy aiming to improve imaging contrast and reduce patient radiation exposure through decoupling of the radioactivity from the targeting vector. The inverse electron-demand Diels-Alder (IEDDA) reaction between a trans-cyclooctene (TCO)-conjugated antibody and a labeled tetrazine holds great promise for pretargeted imaging applications due to its bioorthogonality, rapid kinetics under mild conditions, and formation of stable products. Herein, we describe the use of functionalized carbonylacrylic reagents for site-specific incorporation of TCO onto a human epidermal growth factor receptor 2 (HER2) antibody (THIOMAB) containing an engineered unpaired cysteine residue, generating homogeneous conjugates. Precise labeling of THIOMAB-TCO with a fluorescent or radiolabeled tetrazine revealed the potential of the TCO-functionalized antibody for imaging the HER2 after pretargeting in a cellular context in a HER2 positive breast cancer cell line. Control studies with MDA-MD-231 cells, which do not express HER2, further confirmed the target specificity of the modified antibody. THIOMAB-TCO was also evaluated in vivo after pretargeting and subsequent administration of an 111In-labeled tetrazine. Biodistribution studies in breast cancer tumor-bearing mice showed a significant activity accumulation on HER2+ tumors, which was 2.6-fold higher than in HER2- tumors. Additionally, biodistribution studies with THIOMAB without the TCO handle also resulted in a decreased uptake of 111In-DOTA-Tz on HER2+ tumors. Altogether, these results clearly indicate the occurrence of the click reaction at the tumor site, i.e., pretargeting of SK-BR-3 HER2-expressing cells with THIOMAB-TCO and reaction through the TCO moiety present in the antibody. The combined advantages of site-selectivity and stability of TCO tagged-antibodies could allow application of biorthogonal chemistry strategies for pretargeting imaging with minimal side-reactions and background.
Collapse
Affiliation(s)
- Vera F C Ferreira
- Center for Nuclear Sciences and Technologies (C2TN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| | - Bruno L Oliveira
- Instituto de Medicina Molecular João Lobo Antunes (iMM-JLA), Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Alice D'Onofrio
- Center for Nuclear Sciences and Technologies (C2TN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| | - Carlos M Farinha
- Biosystems and Integrative Sciences Institute (BioISI), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Lurdes Gano
- Center for Nuclear Sciences and Technologies (C2TN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal.,Departamento de Engenharia e Ciências Nucleares (DECN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| | - António Paulo
- Center for Nuclear Sciences and Technologies (C2TN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal.,Departamento de Engenharia e Ciências Nucleares (DECN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| | - Gonçalo J L Bernardes
- Instituto de Medicina Molecular João Lobo Antunes (iMM-JLA), Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal.,Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
| | - Filipa Mendes
- Center for Nuclear Sciences and Technologies (C2TN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal.,Departamento de Engenharia e Ciências Nucleares (DECN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| |
Collapse
|
26
|
Wang Y, Zhang C, Wu H, Feng P. Activation and Delivery of Tetrazine-Responsive Bioorthogonal Prodrugs. Molecules 2020; 25:E5640. [PMID: 33266075 PMCID: PMC7731009 DOI: 10.3390/molecules25235640] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/18/2020] [Accepted: 11/26/2020] [Indexed: 02/05/2023] Open
Abstract
Prodrugs, which remain inert until they are activated under appropriate conditions at the target site, have emerged as an attractive alternative to drugs that lack selectivity and show off-target effects. Prodrugs have traditionally been activated by enzymes, pH or other trigger factors associated with the disease. In recent years, bioorthogonal chemistry has allowed the creation of prodrugs that can be chemically activated with spatio-temporal precision. In particular, tetrazine-responsive bioorthogonal reactions can rapidly activate prodrugs with excellent biocompatibility. This review summarized the recent development of tetrazine bioorthogonal cleavage reaction and great promise for prodrug systems.
Collapse
Affiliation(s)
- Yayue Wang
- Huaxi MR Research Center, Department of Nuclear Medicine, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.W.); (C.Z.)
| | - Chang Zhang
- Huaxi MR Research Center, Department of Nuclear Medicine, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.W.); (C.Z.)
| | - Haoxing Wu
- Huaxi MR Research Center, Department of Nuclear Medicine, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.W.); (C.Z.)
| | - Ping Feng
- Institute of Clinical Trials, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
27
|
Abstract
Over the past decade, theranostic imaging has emerged as a powerful clinical tool in oncology for identifying patients likely to respond to targeted therapies and for monitoring the response of patients to treatment. Herein, we report a theranostic approach to pretargeted radioimmunotherapy (PRIT) based on a pair of radioisotopes of copper: positron-emitting copper-64 (64Cu, t 1/2 = 12.7 h) and beta particle-emitting copper-67 (67Cu, t 1/2 = 61.8 h). This strategy is predicated on the in vivo ligation between a trans-cyclooctene (TCO)-bearing antibody and a tetrazine (Tz)-based radioligand via the rapid and bioorthogonal inverse electron-demand Diels-Alder reaction. Longitudinal therapy studies were conducted in a murine model of human colorectal carcinoma using an immunoconjugate of the huA33 antibody modified with TCO (huA33-TCO) and a 67Cu-labeled Tz radioligand ([67Cu]Cu-MeCOSar-Tz). The injection of huA33-TCO followed 72 h later by the administration of 18.5, 37.0, or 55.5 MBq of [67Cu]Cu-MeCOSar-Tz produced a dose-dependent therapeutic response, with the median survival time increasing from 68 d for the lowest dose to >200 d for the highest. Furthermore, we observed that mice that received the highest dose of [67Cu]Cu-MeCOSar-Tz in a fractionated manner exhibited improved hematological values without sacrificing therapeutic efficacy. Dual radionuclide experiments in which a single administration of huA33-TCO was followed by separate injections of [64Cu]Cu-MeCOSar-Tz and [67Cu]Cu-MeCOSar-Tz revealed that the positron emission tomography images produced by the former accurately predicted the efficacy of the latter. In these experiments, a correlation was observed between the tumoral uptake of [64Cu]Cu-MeCOSar-Tz and the subsequent therapeutic response to [67Cu]Cu-MeCOSar-Tz.
Collapse
|
28
|
Fung K, Sharma SK, Keinänen O, Roche KL, Lewis JS, Zeglis BM. A Molecularly Targeted Intraoperative Near-Infrared Fluorescence Imaging Agent for High-Grade Serous Ovarian Cancer. Mol Pharm 2020; 17:3140-3147. [PMID: 32644804 DOI: 10.1021/acs.molpharmaceut.0c00437] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ovarian cancer is the fifth leading cause of cancer deaths among women, accounting for more deaths than any other cancer of the female reproductive system. The foundation of its management consists of cytoreductive surgery (CRS) followed by systemic chemotherapy, with the completeness of surgical resection consistently identified as one of the most important prognostic factors for the disease. The goal of our investigation is the development of a near-infrared fluorescence (NIRF) imaging agent for the intraoperative imaging of high-grade serous ovarian cancer (HGSOC). As surgeons are currently limited to the visual and manual assessment of tumor tissue during CRS, this technology could facilitate more complete resections as well as serve important functions at other points in the surgical management of the disease. Elevated levels of cancer antigen 125 (CA125) have proven a useful biomarker of HGSOC, and the CA125-targeting antibody B43.13 has shown potential as a platform for immunoPET imaging in murine models of ovarian cancer. Herein, we report the development of a NIRF imaging agent based on B43.13: ssB43.13-IR800. We site-specifically modified the heavy chain glycans of B43.13 with the near-infrared dye IRDye 800CW using a chemoenzymatic approach developed in our laboratories. SDS-PAGE analysis confirmed the specificity of the conjugation reaction, and flow cytometry, immunostaining, and fluorescence microscopy verified the specific binding of ssB43.13-IR800 to CA125-expressing OVCAR3 human ovarian cancer cells. NIRF imaging studies demonstrated that ssB43.13-IR800 can be used to image CA125-expressing HGSOC tumors in subcutaneous, orthotopic, and patient-derived xenograft mouse models. Finally, ex vivo analyses confirmed that ssB43.13-IR800 can bind and identify CA125-expressing cells in primary tumor and metastatic lymph node samples from human patients with HGSOC.
Collapse
Affiliation(s)
- Kimberly Fung
- Department of Chemistry, Hunter College, City University of New York, New York, New York 10021, United States.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
| | | | - Outi Keinänen
- Department of Chemistry, Hunter College, City University of New York, New York, New York 10021, United States
| | - Kara Long Roche
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | | | - Brian M Zeglis
- Department of Chemistry, Hunter College, City University of New York, New York, New York 10021, United States.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
| |
Collapse
|
29
|
Abstract
Immuno-positron emission tomography (immunoPET) is a paradigm-shifting molecular imaging modality combining the superior targeting specificity of monoclonal antibody (mAb) and the inherent sensitivity of PET technique. A variety of radionuclides and mAbs have been exploited to develop immunoPET probes, which has been driven by the development and optimization of radiochemistry and conjugation strategies. In addition, tumor-targeting vectors with a short circulation time (e.g., Nanobody) or with an enhanced binding affinity (e.g., bispecific antibody) are being used to design novel immunoPET probes. Accordingly, several immunoPET probes, such as 89Zr-Df-pertuzumab and 89Zr-atezolizumab, have been successfully translated for clinical use. By noninvasively and dynamically revealing the expression of heterogeneous tumor antigens, immunoPET imaging is gradually changing the theranostic landscape of several types of malignancies. ImmunoPET is the method of choice for imaging specific tumor markers, immune cells, immune checkpoints, and inflammatory processes. Furthermore, the integration of immunoPET imaging in antibody drug development is of substantial significance because it provides pivotal information regarding antibody targeting abilities and distribution profiles. Herein, we present the latest immunoPET imaging strategies and their preclinical and clinical applications. We also emphasize current conjugation strategies that can be leveraged to develop next-generation immunoPET probes. Lastly, we discuss practical considerations to tune the development and translation of immunoPET imaging strategies.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
| | - Zachary T Rosenkrans
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Quan-Yong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53705, United States
| |
Collapse
|
30
|
Imberti C, Adumeau P, Blower JE, Al Salemee F, Baguña Torres J, Lewis JS, Zeglis BM, Terry SYA, Blower PJ. Manipulating the In Vivo Behaviour of 68Ga with Tris(Hydroxypyridinone) Chelators: Pretargeting and Blood Clearance. Int J Mol Sci 2020; 21:E1496. [PMID: 32098299 PMCID: PMC7073083 DOI: 10.3390/ijms21041496] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 02/07/2023] Open
Abstract
Pretargeting is widely explored in immunoPET as a strategy to reduce radiation exposure of non-target organs and allow the use of short-lived radionuclides that would not otherwise be compatible with the slow pharmacokinetic profiles of antibodies. Here we investigate a pretargeting strategy based on gallium-68 and the chelator THPMe as a high-affinity pair capable of combining in vivo. After confirming the ability of THPMe to bind 68Ga in vivo at low concentrations, the bifunctional THPMe-NCS was conjugated to a humanised huA33 antibody targeting the A33 glycoprotein. Imaging experiments performed in nude mice bearing A33-positive SW1222 colorectal cancer xenografts compared pretargeting (100 μg of THPMe-NCS-huA33, followed after 24 h by 8-10 MBq of 68Ga3+) with both a directly labelled radioimmunoconjugate (89Zr-DFO-NCS-huA33, 88 μg, 7 MBq) and a 68Ga-only negative control (8-10 MBq of 68Ga3+). Imaging was performed 25 h after antibody administration (1 h after 68Ga3+ administration for negative control). No difference between pretargeting and the negative control was observed, suggesting that pretargeting via metal chelation is not feasible using this model. However, significant accumulation of "unchelated" 68Ga3+ in the tumour was found (12.9 %ID/g) even without prior administration of THPMe-NCS-huA33, though tumour-to-background contrast was impaired by residual activity in the blood. Therefore, the 68Ga-only experiment was repeated using THPMe (20 μg, 1 h after 68Ga3+ administration) to clear circulating 68Ga3+, producing a three-fold improvement of the tumour-to-blood activity concentration ratio. Although preliminary, these results highlight the potential of THPMe as a 68Ga clearing agent in imaging applications with gallium citrate.
Collapse
Affiliation(s)
- Cinzia Imberti
- School of Biomedical Engineering and Imaging Sciences, King’s College London, Fourth Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK
| | - Pierre Adumeau
- Department of Chemistry, Hunter College, City University of New York, New York, NY 10021, USA
| | - Julia E. Blower
- School of Biomedical Engineering and Imaging Sciences, King’s College London, Fourth Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK
| | - Fahad Al Salemee
- School of Biomedical Engineering and Imaging Sciences, King’s College London, Fourth Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK
| | - Julia Baguña Torres
- School of Biomedical Engineering and Imaging Sciences, King’s College London, Fourth Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK
| | - Jason S. Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Brian M. Zeglis
- Department of Chemistry, Hunter College, City University of New York, New York, NY 10021, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Samantha Y. A. Terry
- School of Biomedical Engineering and Imaging Sciences, King’s College London, Fourth Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK
| | - Philip J. Blower
- School of Biomedical Engineering and Imaging Sciences, King’s College London, Fourth Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK
| |
Collapse
|
31
|
Toftevall H, Nyhlén H, Olsson F, Sjögren J. Antibody Conjugations via Glycosyl Remodeling. Methods Mol Biol 2020; 2078:131-145. [PMID: 31643054 DOI: 10.1007/978-1-4939-9929-3_9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The antibody Fc-glycans are interesting targets for conjugation of cytotoxic compounds due to their localization and their chemical composition. In striving to obtain site-specific conjugates, the antibody Fc-glycans have been explored in numerous ways. Here we present a two-step enzymatic methodology coupled to click-chemistry for conjugation at the core GlcNAc of the Fc-glycan resulting in ADCs that are homogenous with a DAR 2.0, retain antigen binding, and display cytotoxic anti-tumor effects both in vitro and in vivo.
Collapse
|
32
|
Recent Advances in Bioorthogonal Click Chemistry for Efficient Synthesis of Radiotracers and Radiopharmaceuticals. Molecules 2019; 24:molecules24193567. [PMID: 31581645 PMCID: PMC6803924 DOI: 10.3390/molecules24193567] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/23/2019] [Accepted: 09/27/2019] [Indexed: 12/17/2022] Open
Abstract
In recent years, several catalyst-free site-specific reactions have been investigated for the efficient conjugation of biomolecules, nanomaterials, and living cells. Representative functional group pairs for these reactions include the following: (1) azide and cyclooctyne for strain-promoted cycloaddition reaction, (2) tetrazine and trans-alkene for inverse-electron-demand-Diels–Alder reaction, and (3) electrophilic heterocycles and cysteine for rapid condensation/addition reaction. Due to their excellent specificities and high reaction rates, these conjugation methods have been utilized for the labeling of radioisotopes (e.g., radiohalogens, radiometals) to various target molecules. The radiolabeled products prepared by these methods have been applied to preclinical research, such as in vivo molecular imaging, pharmacokinetic studies, and radiation therapy of cancer cells. In this review, we explain the basics of these chemical reactions and introduce their recent applications in the field of radiopharmacy and chemical biology. In addition, we discuss the significance, current challenges, and prospects of using bioorthogonal conjugation reactions.
Collapse
|
33
|
Marculescu C, Lakshminarayanan A, Gault J, Knight JC, Folkes LK, Spink T, Robinson CV, Vallis K, Davis BG, Cornelissen B. Probing the limits of Q-tag bioconjugation of antibodies. Chem Commun (Camb) 2019; 55:11342-11345. [PMID: 31479092 PMCID: PMC6788405 DOI: 10.1039/c9cc02303h] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 07/25/2019] [Indexed: 12/25/2022]
Abstract
Site-selective labelling of antibodies (Abs) can circumvent problems from heterogeneity of conventional conjugation. Here, we evaluate the industrially-applied chemoenzymatic 'Q-tag' strategy based on transglutaminase-mediated (TGase) amide-bond formation in the generation of 89Zr-radiolabelled antibody conjugates. We show that, despite previously suggested high regioselectivity of TGases, in the anti-Her2 Ab Herceptin™ more precise native MS indicates only 70-80% functionalization at the target site (Q298H), in competition with modification at other sites, such as Q3H critically close to the CDR1 region.
Collapse
Affiliation(s)
- Cristina Marculescu
- CRUK/MRC Oxford Institute for Radiation Oncology
, Department of Oncology, University of Oxford
,
Oxford
, OX3 7DQ
, UK
.
;
- Chemistry Research Laboratory
, University of Oxford
,
Oxford
, OX1 3TA
, UK
.
| | - Abirami Lakshminarayanan
- CRUK/MRC Oxford Institute for Radiation Oncology
, Department of Oncology, University of Oxford
,
Oxford
, OX3 7DQ
, UK
.
;
- Chemistry Research Laboratory
, University of Oxford
,
Oxford
, OX1 3TA
, UK
.
| | - Joseph Gault
- Chemistry Research Laboratory
, University of Oxford
,
Oxford
, OX1 3TA
, UK
.
| | - James C. Knight
- CRUK/MRC Oxford Institute for Radiation Oncology
, Department of Oncology, University of Oxford
,
Oxford
, OX3 7DQ
, UK
.
;
| | - Lisa K. Folkes
- CRUK/MRC Oxford Institute for Radiation Oncology
, Department of Oncology, University of Oxford
,
Oxford
, OX3 7DQ
, UK
.
;
| | - Thomas Spink
- CRUK/MRC Oxford Institute for Radiation Oncology
, Department of Oncology, University of Oxford
,
Oxford
, OX3 7DQ
, UK
.
;
| | - Carol V. Robinson
- Chemistry Research Laboratory
, University of Oxford
,
Oxford
, OX1 3TA
, UK
.
| | - Katherine Vallis
- CRUK/MRC Oxford Institute for Radiation Oncology
, Department of Oncology, University of Oxford
,
Oxford
, OX3 7DQ
, UK
.
;
| | - Benjamin G. Davis
- Chemistry Research Laboratory
, University of Oxford
,
Oxford
, OX1 3TA
, UK
.
| | - Bart Cornelissen
- CRUK/MRC Oxford Institute for Radiation Oncology
, Department of Oncology, University of Oxford
,
Oxford
, OX3 7DQ
, UK
.
;
| |
Collapse
|
34
|
Keinänen O, Brennan JM, Membreno R, Fung K, Gangangari K, Dayts EJ, Williams CJ, Zeglis BM. Dual Radionuclide Theranostic Pretargeting. Mol Pharm 2019; 16:4416-4421. [PMID: 31483993 DOI: 10.1021/acs.molpharmaceut.9b00746] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recent years have played witness to the advent of nuclear theranostics: the synergistic use of "matched pair" radiopharmaceuticals for diagnostic imaging and targeted radiotherapy. In this investigation, we report the extension of this concept to in vivo pretargeting based on the rapid and bioorthogonal inverse electron demand Diels-Alder reaction between tetrazine (Tz) and trans-cyclooctene (TCO). We demonstrate that a single injection of a TCO-modified immunoconjugate can be used as a platform for pretargeted PET imaging and radiotherapy via the sequential administration of a pair of Tz-bearing radioligands labeled with the positron-emitting radiometal copper-64 (t1/2 ≈ 12.7 h) and the beta-emitting radiometal lutetium-177 (t1/2 ≈ 6.7 days). More specifically, a mouse model of human colorectal carcinoma received a dose of the A33 antigen-targeting immunoconjugate huA33-TCO, followed 24 and 48 h later by injections of [64Cu]Cu-SarAr-Tz and [177Lu]Lu-DOTA-PEG7-Tz, respectively. This approach produces high activity concentrations of both radioligands in tumor tissue (16.4 ± 2.7 %ID/g for [64Cu]Cu-SarAr-Tz at 48 h post-injection and 18.1 ± 2.1 %ID/g for [177Lu]Lu-DOTA-PEG7-Tz at 120 h post-injection) as well as promising tumor-to-healthy organ activity concentration ratios. Ultimately, we believe that this work could not only have important implications in nuclear theranostics-most excitingly with isotopologue-based radioligand pairs such as [64Cu]Cu-SarAr-Tz and [67Cu]Cu-SarAr-Tz-but also in the delivery of fractionated doses during pretargeted radioimmunotherapy.
Collapse
Affiliation(s)
- Outi Keinänen
- Department of Chemistry , Hunter College, City University of New York , New York , New York 10021 , United States
| | - James M Brennan
- Department of Chemistry , Hunter College, City University of New York , New York , New York 10021 , United States
| | - Rosemery Membreno
- Department of Chemistry , Hunter College, City University of New York , New York , New York 10021 , United States.,Ph.D. Program in Chemistry , The Graduate Center of the City University of New York , New York , New York 10016 , United States
| | - Kimberly Fung
- Department of Chemistry , Hunter College, City University of New York , New York , New York 10021 , United States.,Ph.D. Program in Chemistry , The Graduate Center of the City University of New York , New York , New York 10016 , United States
| | - Kishore Gangangari
- Department of Chemistry , Hunter College, City University of New York , New York , New York 10021 , United States.,Ph.D. Program in Chemistry , The Graduate Center of the City University of New York , New York , New York 10016 , United States
| | - Eric J Dayts
- Department of Chemistry , Hunter College, City University of New York , New York , New York 10021 , United States
| | - Carter J Williams
- Department of Chemistry , Hunter College, City University of New York , New York , New York 10021 , United States
| | - Brian M Zeglis
- Department of Chemistry , Hunter College, City University of New York , New York , New York 10021 , 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 , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States.,Department of Radiology , Weill Cornell Medical College , New York , New York 10065 , United States
| |
Collapse
|
35
|
Membreno R, Keinänen OM, Cook BE, Tully KM, Fung KC, Lewis JS, Zeglis BM. Toward the Optimization of Click-Mediated Pretargeted Radioimmunotherapy. Mol Pharm 2019; 16:2259-2263. [PMID: 30912951 DOI: 10.1021/acs.molpharmaceut.9b00062] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Pretargeted radioimmunotherapy (PRIT) based on the inverse electron demand Diels-Alder reaction has shown promise in murine models of disease, yet the radiation dosimetry of this approach must be optimized to make it a viable clinical option. To this end, we have leveraged two recent developments in pretargeted imaging-dendritic scaffolds and masking agents-to improve the dosimetric profile of a proof-of-concept PRIT system that is based on the huA33 antibody, a 177Lu-labeled tetrazine radioligand ([177Lu]Lu-DOTA-PEG7-Tz), and a mouse model of A33 antigen-expressing colorectal carcinoma. Pretargeting using an huA33 immunoconjugate bearing a trans-cyclooctene-decorated dendritic scaffold (sshuA33-DEN-TCO) produced significantly higher tumoral activity concentrations at 120 h post-injection (23.0 ± 2.2 %ID/g) than those achieved with an analogous, dendrimer-lacking immunoconjugate (12.7 ± 2.6 %ID/g). However, pretargeting using sshuA33-DEN-TCO also resulted in increased activity concentrations in the blood at the same time point (1.9 ± 0.4 %ID/g) compared to the dendrimer-lacking construct (0.7 ± 0.2 %ID/g), thereby curtailing improvements to the tumor-to-blood therapeutic ratio of the system. In order to circumvent this issue, a tetrazine-labeled, dextran-based masking agent (Tz-DP) was injected prior to the radioligand to prevent the ligation between [177Lu]Lu-DOTA-PEG7-Tz and circulating immunoconjugate. This approach dramatically decreased the absorbed dose to the blood but also attenuated the absorbed dose to the tumor and increased the absorbed dose to the lungs. Ultimately, these data suggest that dendritic scaffolds and masking agents could be used to improve the dosimetry of PRIT, but the combination of these technologies will require extensive optimization.
Collapse
Affiliation(s)
- Rosemery Membreno
- Department of Chemistry , Hunter College of the City University of New York , New York , New York 10021 , United States.,Ph.D. Program in Chemistry , The Graduate Center of the City University of New York , New York , New York 10016 , United States
| | - Outi M Keinänen
- Department of Chemistry , Hunter College of the City University of New York , New York , New York 10021 , United States
| | - Brendon E Cook
- Department of Chemistry , Hunter College of the City University of New York , New York , New York 10021 , United States.,Ph.D. Program in Chemistry , The Graduate Center of the City University of New York , New York , New York 10016 , United States
| | | | - Kimberly C Fung
- Department of Chemistry , Hunter College of the City University of New York , New York , New York 10021 , United States.,Ph.D. Program in Chemistry , The Graduate Center of the City University of New York , New York , New York 10016 , United States
| | | | - Brian M Zeglis
- Department of Chemistry , Hunter College of the City University of New York , New York , New York 10021 , United States.,Ph.D. Program in Chemistry , The Graduate Center of the City University of New York , New York , New York 10016 , United States
| |
Collapse
|
36
|
Pellico J, Fernández-Barahona I, Benito M, Gaitán-Simón Á, Gutiérrez L, Ruiz-Cabello J, Herranz F. Unambiguous detection of atherosclerosis using bioorthogonal nanomaterials. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 17:26-35. [DOI: 10.1016/j.nano.2018.12.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 12/13/2018] [Accepted: 12/28/2018] [Indexed: 12/21/2022]
|
37
|
Vivier D, Sharma SK, Adumeau P, Rodriguez C, Fung K, Zeglis BM. The Impact of FcγRI Binding on Immuno-PET. J Nucl Med 2019; 60:1174-1182. [PMID: 30733320 DOI: 10.2967/jnumed.118.223636] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 01/01/2018] [Indexed: 01/07/2023] Open
Abstract
Antibodies are promising vectors for PET imaging. However, the high uptake of radioimmunoconjugates in nontarget tissues such as the liver and spleen hampers their performance as radiotracers. This off-target uptake can lead to suboptimal tumor-to-background activity concentration ratios, decreasing the contrast of images and reducing their diagnostic utility. A possible cause of this uptake is the sequestration of radioimmunoconjugates by immune cells bearing Fc-γ-receptors (FcγR) that bind to the Fc regions of antibodies. Methods: Since the heavy chain glycans influence the affinity of FcγR for the Fc domain, we set out to investigate whether radioimmunoconjugates with truncated glycans would exhibit altered binding to FcγRI and, in turn, improved in vivo performance. Using the HER2-targeting antibody trastuzumab, we synthesized a series of desferrioxamine-bearing immunoconjugates with differing glycosylation states and interrogated their FcγRI binding via surface plasmon resonance, enzyme-linked immunosorbent assay, and flow cytometry. Furthermore, we labeled these immunoconjugates with 89Zr and explored their biodistribution in athymic nude, NSG, and humanized NSG mice bearing human epidermal growth factor receptor 2-expressing human breast cancer xenografts. Results: We observed a strong correlation between the impaired in vitro FcγRI binding of deglycosylated immunoconjugates and significant decreases in the in vivo off-target uptake of the corresponding 89Zr-labeled radioimmunoconjugates (i.e., liver activity concentrations are reduced by ∼3.5-fold in humanized NSG mice). These reductions in off-target uptake were accompanied by concomitant increases in the tumoral activity concentrations of the glycoengineered radioimmunoconjugates, ultimately yielding improved tumor-to-healthy organ contrast and higher quality PET images. Conclusion: Our findings suggest that the deglycosylation of antibodies represents a facile strategy for improving the quality of immuno-PET in animal models as well as in certain patient populations.
Collapse
Affiliation(s)
- Delphine Vivier
- Department of Chemistry, Hunter College of the City University of New York, New York, New York
| | - Sai Kiran Sharma
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Pierre Adumeau
- Department of Chemistry, Hunter College of the City University of New York, New York, New York
| | - Cindy Rodriguez
- Department of Chemistry, Hunter College of the City University of New York, New York, New York
| | - Kimberly Fung
- Department of Chemistry, Hunter College of the City University of New York, New York, New York.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, New York
| | - Brian M Zeglis
- Department of Chemistry, Hunter College of the City University of New York, New York, New York .,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York; and.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, New York
| |
Collapse
|
38
|
Roche M, Specklin S, Richard M, Hinnen F, Génermont K, Kuhnast B. [ 18 F]FPyZIDE: A versatile prosthetic reagent for the fluorine-18 radiolabeling of biologics via copper-catalyzed or strain-promoted alkyne-azide cycloadditions. J Labelled Comp Radiopharm 2019; 62:95-108. [PMID: 30556584 DOI: 10.1002/jlcr.3701] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/07/2018] [Accepted: 12/11/2018] [Indexed: 11/05/2022]
Abstract
Methods for the radiolabeling of biologics with fluorine-18 have been of interest for several decades. A common approach consists in the preparation of a prosthetic reagent, a small molecule bearing a fluorine-18 that is conjugated with the macromolecule to an appropriate function. Click chemistry, and more particularly cycloadditions, is an interesting approach to radiolabel molecules thanks to mild reaction conditions, high yields, low by-products formation, and strong orthogonality. Moreover, the chemical functions involved in the cycloaddition reaction are stable in the drastic radiofluorination conditions, thus allowing a simple radiosynthetic route to prepare the prosthetic reagent. We report herein the radiosynthesis of 18 F-FPyZIDE, a pyridine-based azide-bearing prosthetic reagent. We exemplified its conjugation via copper-catalyzed cycloaddition (CuAAC) and strain-promoted cycloaddition (SPAAC) with several terminal alkyne or strained alkyne model compounds.
Collapse
Affiliation(s)
- Mélanie Roche
- IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Simon Specklin
- IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Mylène Richard
- IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Françoise Hinnen
- IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Kevin Génermont
- IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Bertrand Kuhnast
- IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| |
Collapse
|
39
|
Richard M, Truillet C, Tran VL, Liu H, Porte K, Audisio D, Roche M, Jego B, Cholet S, Fenaille F, Kuhnast B, Taran F, Specklin S. New fluorine-18 pretargeting PET imaging by bioorthogonal chlorosydnone–cycloalkyne click reaction. Chem Commun (Camb) 2019; 55:10400-10403. [DOI: 10.1039/c9cc05486c] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A PET pretargeting approach using strain-promoted sydnone–alkyne cycloaddition.
Collapse
Affiliation(s)
- Mylène Richard
- UMR 1023 IMIV
- Service Hospitalier Frédéric Joliot (SHFJ)
- CEA, Inserm
- Université Paris Sud, CNRS
- Université Paris-Saclay
| | - Charles Truillet
- UMR 1023 IMIV
- Service Hospitalier Frédéric Joliot (SHFJ)
- CEA, Inserm
- Université Paris Sud, CNRS
- Université Paris-Saclay
| | - Vu Long Tran
- UMR 1023 IMIV
- Service Hospitalier Frédéric Joliot (SHFJ)
- CEA, Inserm
- Université Paris Sud, CNRS
- Université Paris-Saclay
| | - Hui Liu
- Service de Chimie Bio-organique et Marquage DRF-JOLIOT-SCBM
- CEA, Université Paris-Saclay
- 91191 Gif-sur-Yvette
- France
| | - Karine Porte
- Service de Chimie Bio-organique et Marquage DRF-JOLIOT-SCBM
- CEA, Université Paris-Saclay
- 91191 Gif-sur-Yvette
- France
| | - Davide Audisio
- Service de Chimie Bio-organique et Marquage DRF-JOLIOT-SCBM
- CEA, Université Paris-Saclay
- 91191 Gif-sur-Yvette
- France
| | - Mélanie Roche
- UMR 1023 IMIV
- Service Hospitalier Frédéric Joliot (SHFJ)
- CEA, Inserm
- Université Paris Sud, CNRS
- Université Paris-Saclay
| | - Benoit Jego
- UMR 1023 IMIV
- Service Hospitalier Frédéric Joliot (SHFJ)
- CEA, Inserm
- Université Paris Sud, CNRS
- Université Paris-Saclay
| | - Sophie Cholet
- Service de Pharmacologie et d’Immunoanalyse (SPI)
- CEA/DRF/JOLIOT
- Université Paris Saclay
- F-91191 Gif-sur-Yvette
- France
| | - François Fenaille
- Service de Pharmacologie et d’Immunoanalyse (SPI)
- CEA/DRF/JOLIOT
- Université Paris Saclay
- F-91191 Gif-sur-Yvette
- France
| | - Bertrand Kuhnast
- UMR 1023 IMIV
- Service Hospitalier Frédéric Joliot (SHFJ)
- CEA, Inserm
- Université Paris Sud, CNRS
- Université Paris-Saclay
| | - Frédéric Taran
- Service de Chimie Bio-organique et Marquage DRF-JOLIOT-SCBM
- CEA, Université Paris-Saclay
- 91191 Gif-sur-Yvette
- France
| | - Simon Specklin
- UMR 1023 IMIV
- Service Hospitalier Frédéric Joliot (SHFJ)
- CEA, Inserm
- Université Paris Sud, CNRS
- Université Paris-Saclay
| |
Collapse
|
40
|
Allen KJH, Jiao R, Malo ME, Frank C, Dadachova E. Biodistribution of a Radiolabeled Antibody in Mice as an Approach to Evaluating Antibody Pharmacokinetics. Pharmaceutics 2018; 10:pharmaceutics10040262. [PMID: 30563123 PMCID: PMC6320949 DOI: 10.3390/pharmaceutics10040262] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/21/2018] [Accepted: 12/01/2018] [Indexed: 12/14/2022] Open
Abstract
(1) Background: Monoclonal antibodies are used in the treatment of multiple conditions including cancer, autoimmune disorders, and infectious diseases. One of the initial steps in the selection of an antibody candidate for further pre-clinical development is determining its pharmacokinetics in small animal models. The use of mass spectrometry and other techniques to determine the fate of these antibodies is laborious and expensive. Here we describe a straightforward and highly reproducible methodology for utilizing radiolabeled antibodies for pharmacokinetics studies. (2) Methods: Commercially available bifunctional linker CHXA” and 111Indium radionuclide were used. A melanin-specific chimeric antibody A1 and an isotype matching irrelevant control A2 were conjugated with the CHXA”, and then radiolabeled with 111In. The biodistribution was performed at 4 and 24 h time points in melanoma tumor-bearing and healthy C57BL/6 female mice. (3) The biodistribution of the melanin-binding antibody showed the significant uptake in the tumor, which increased with time, and very low uptake in healthy melanin-containing tissues such as the retina of the eye and melanized skin. This biodistribution pattern in healthy tissues was very close to that of the isotype matching control antibody. (4) Conclusions: The biodistribution experiment allows us to assess the pharmacokinetics of both antibodies side by side and to make a conclusion regarding the suitability of specific antibodies for further development.
Collapse
Affiliation(s)
- Kevin J H Allen
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| | - Rubin Jiao
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| | - Mackenzie E Malo
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| | - Connor Frank
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| |
Collapse
|
41
|
Litau S, Seibold U, Wängler B, Schirrmacher R, Wängler C. iEDDA Conjugation Reaction in Radiometal Labeling of Peptides with 68Ga and 64Cu: Unexpected Findings. ACS OMEGA 2018; 3:14039-14053. [PMID: 30411057 PMCID: PMC6217686 DOI: 10.1021/acsomega.8b01926] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/15/2018] [Indexed: 05/31/2023]
Abstract
The inverse electron demand Diels-Alder conjugation reaction has gained increasing importance over the past few years for efficient in vivo and ex vivo radiometal labeling of antibodies. However, the application of this very fast reaction type has not been studied for radiolabeling of peptides so far. We show here the synthesis of 3-benzyl-1,2,4,5-tetrazine-comprising ((1,4,7,10-tetraazacyclododecane-4,7,10-triyl)triacetic acid-1-glutaric acid) (DOTA-GA) and ((1,4,7-triazacyclononane-4,7-diyl)diacetic acid-1-glutaric acid) (NODA-GA) chelators and their radiometal labeling with 68Ga3+ and 64Cu2+. The secondary labeling precursors 68Ga-DOTA-GA-Tz, 68Ga-NODA-GA-Tz, and 64Cu-DOTA-GA-Tz were obtained in high radiochemical yields (RCYs) and purities as well as molar activities for further labeling of trans-cyclooctene (TCO)-modified peptides. However, the following reactions of the radiometal-labeled tetrazines with different TCO-comprising model peptide analogs unexpectedly resulted in the formation of a considerable amount of side products (20-55%) which limits the overall achievable RCYs and purities as well as molar activities of the target radiopeptides. Under otherwise identical, nonradioactive reaction conditions, this effect could however not be observed. In contrast, the corresponding one-step radiolabeling protocols provided the target 68Ga-labeled radiopeptides in exceptionally high RCYs and purities of ≥99% and molar activities of 68-72 GBq/μmol starting from activities of 340-358 MBq of 68Ga. Thus, the usefulness of the two-step labeling of TCO-modified peptides with radiometal-labeled chelator-tetrazines seems to be limited.
Collapse
Affiliation(s)
- Shanna Litau
- Department
of Clinical Radiology and Nuclear Medicine, Biomedical
Chemistry and Department of Clinical Radiology and Nuclear Medicine, Molecular
Imaging and Radiochemistry, Medical Faculty
Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim 68167, Germany
| | - Uwe Seibold
- Department
of Clinical Radiology and Nuclear Medicine, Biomedical
Chemistry and Department of Clinical Radiology and Nuclear Medicine, Molecular
Imaging and Radiochemistry, Medical Faculty
Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim 68167, Germany
| | - Björn Wängler
- Department
of Clinical Radiology and Nuclear Medicine, Biomedical
Chemistry and Department of Clinical Radiology and Nuclear Medicine, Molecular
Imaging and Radiochemistry, Medical Faculty
Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim 68167, Germany
| | - Ralf Schirrmacher
- Department
of Oncology, Division of Oncological Imaging, University of Alberta, 11560 University Avenue, Edmonton T6G 1Z2, Alberta, Canada
| | - Carmen Wängler
- Department
of Clinical Radiology and Nuclear Medicine, Biomedical
Chemistry and Department of Clinical Radiology and Nuclear Medicine, Molecular
Imaging and Radiochemistry, Medical Faculty
Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim 68167, Germany
| |
Collapse
|
42
|
Pretargeted Imaging with Gallium-68-Improving the Binding Capability by Increasing the Number of Tetrazine Motifs. Pharmaceuticals (Basel) 2018; 11:ph11040102. [PMID: 30314332 PMCID: PMC6316846 DOI: 10.3390/ph11040102] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 01/29/2023] Open
Abstract
The inverse electron-demand Diels-Alder reaction between 1,2,4,5-tetrazine (Tz) and trans-cyclooct-2-ene (TCO) has gained increasing attraction among extensive studies on click chemistry due to its exceptionally fast reaction kinetics and high selectivity for in vivo pretargeting applications including PET imaging. The facile two-step approach utilizing TCO-modified antibodies as targeting structures has not made it into clinics yet. An increase in the blood volume of humans in comparison to mice seems to be the major limitation. This study aims to show if the design of multimeric Tz-ligands by chelator scaffolding can improve the binding capacity and may lead to enhanced PET imaging with gallium-68. We utilized for this purpose the macrocyclic siderophore Fusarinine C (FSC) which allows conjugation of up to three Tz-residues due to three primary amines available for site specific modification. The resulting mono- di- and trimeric conjugates were radiolabelled with gallium-68 and characterized in vitro (logD, protein binding, stability, binding towards TCO modified rituximab (RTX)) and in vivo (biodistribution- and imaging studies in normal BALB/c mice using a simplified RTX-TCO tumour surrogate). The 68Ga-labelled FSC-based Tz-ligands showed suitable hydrophilicity, high stability and high targeting specificity. The binding capacity to RTX-TCO was increased according to the grade of multimerization. Corresponding in vivo studies showed a multimerization typical profile but generally suitable pharmacokinetics with low accumulation in non-targeted tissue. Imaging studies in RTX-TCO tumour surrogate bearing BALB/c mice confirmed this trend and revealed improved targeting by multimerization as increased accumulation in RTX-TCO positive tissue was observed.
Collapse
|
43
|
Mandikian D, Rafidi H, Adhikari P, Venkatraman P, Nazarova L, Fung G, Figueroa I, Ferl GZ, Ulufatu S, Ho J, McCaughey C, Lau J, Yu SF, Prabhu S, Sadowsky J, Boswell CA. Site-specific conjugation allows modulation of click reaction stoichiometry for pretargeted SPECT imaging. MAbs 2018; 10:1269-1280. [PMID: 30199303 PMCID: PMC6284555 DOI: 10.1080/19420862.2018.1521132] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Antibody pretargeting is a promising strategy for improving molecular imaging, wherein the separation in time of antibody targeting and radiolabeling can lead to rapid attainment of high contrast, potentially increased sensitivity, and reduced patient radiation exposure. The inverse electron demand Diels-Alder ‘click’ reaction between trans-cyclooctene (TCO) conjugated antibodies and radiolabeled tetrazines presents an ideal platform for pretargeted imaging due to rapid reaction kinetics, bioorthogonality, and potential for optimization of both slow and fast clearing components. Herein, we evaluated a series of anti-human epidermal growth factor receptor 2 (HER2) pretargeting antibodies containing distinct molar ratios of site-specifically incorporated TCO. The effect of stoichiometry on tissue distribution was assessed for pretargeting TCO-modified antibodies (monitored by 125I) and subsequent accumulation of an 111In-labeled tetrazine in a therapeutically relevant HER2+tumor-bearing mouse model. Single photon emission computed tomography (SPECT) imaging was also employed to assess tumor imaging at various TCO-to-monoclonal antibody (mAb) ratios. Increasing TCO-to-mAb molar ratios correlated with increased in vivo click reaction efficiency evident by increased tumor distribution and systemic exposure of 111In-labeled tetrazines. The pharmacokinetics of TCO-modified antibodies did not vary with stoichiometry. Pretargeted SPECT imaging of HER2-expressing tumors using 111In-labeled tetrazine demonstrated robust click reaction with circulating antibody at ~2 hours and good tumor delineation for both the 2 and 6 TCO-to-mAb ratio variants at 24 hours, consistent with a limited cell-surface pool of pretargeted antibody and benefit from further distribution and internalization. To our knowledge, this represents the first reported systematic analysis of how pretargeted imaging is affected solely by variation in click reaction stoichiometry through site-specific conjugation chemistry.
Collapse
Affiliation(s)
- Danielle Mandikian
- a Preclinical and Translational Pharmacokinetics , Genentech Inc ., South San Francisco , CA , USA
| | - Hanine Rafidi
- a Preclinical and Translational Pharmacokinetics , Genentech Inc ., South San Francisco , CA , USA
| | - Pragya Adhikari
- b Protein Chemistry , Genentech Inc ., South San Francisco , CA , USA
| | - Priya Venkatraman
- a Preclinical and Translational Pharmacokinetics , Genentech Inc ., South San Francisco , CA , USA
| | - Lidia Nazarova
- a Preclinical and Translational Pharmacokinetics , Genentech Inc ., South San Francisco , CA , USA
| | - Gabriel Fung
- a Preclinical and Translational Pharmacokinetics , Genentech Inc ., South San Francisco , CA , USA
| | - Isabel Figueroa
- a Preclinical and Translational Pharmacokinetics , Genentech Inc ., South San Francisco , CA , USA
| | - Gregory Z Ferl
- a Preclinical and Translational Pharmacokinetics , Genentech Inc ., South San Francisco , CA , USA
| | - Sheila Ulufatu
- c In Vivo Studies , Genentech Research and Early Development , South San Francisco , CA , USA
| | - Jason Ho
- c In Vivo Studies , Genentech Research and Early Development , South San Francisco , CA , USA
| | - Cynthia McCaughey
- c In Vivo Studies , Genentech Research and Early Development , South San Francisco , CA , USA
| | - Jeffrey Lau
- d Translational Oncology , Genentech Inc ., South San Francisco , CA , USA
| | - Shang-Fan Yu
- d Translational Oncology , Genentech Inc ., South San Francisco , CA , USA
| | - Saileta Prabhu
- a Preclinical and Translational Pharmacokinetics , Genentech Inc ., South San Francisco , CA , USA
| | - Jack Sadowsky
- b Protein Chemistry , Genentech Inc ., South San Francisco , CA , USA
| | - C Andrew Boswell
- a Preclinical and Translational Pharmacokinetics , Genentech Inc ., South San Francisco , CA , USA
| |
Collapse
|
44
|
Ahmedova A, Todorov B, Burdzhiev N, Goze C. Copper radiopharmaceuticals for theranostic applications. Eur J Med Chem 2018; 157:1406-1425. [DOI: 10.1016/j.ejmech.2018.08.051] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 08/15/2018] [Accepted: 08/18/2018] [Indexed: 12/12/2022]
|
45
|
Dmochowska N, Wardill HR, Hughes PA. Advances in Imaging Specific Mediators of Inflammatory Bowel Disease. Int J Mol Sci 2018; 19:ijms19092471. [PMID: 30134572 PMCID: PMC6164364 DOI: 10.3390/ijms19092471] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/02/2018] [Accepted: 08/20/2018] [Indexed: 12/14/2022] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by chronic remitting and relapsing inflammation of the lower gastrointestinal tract. The etiology underlying IBD remains unknown, but it is thought to involve a hypersensitive immune response to environmental antigens, including the microbiota. Diagnosis and monitoring of IBD is heavily reliant on endoscopy, which is invasive and does not provide information regarding specific mediators. This review describes recent developments in imaging of IBD with a focus on positron emission tomography (PET) and single-photon emission computed tomography (SPECT) of inflammatory mediators, and how these developments may be applied to the microbiota.
Collapse
Affiliation(s)
- Nicole Dmochowska
- Centre for Nutrition and GI Diseases, Adelaide Medical School, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide 5000, Australia.
| | - Hannah R Wardill
- Centre for Nutrition and GI Diseases, Adelaide Medical School, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide 5000, Australia.
| | - Patrick A Hughes
- Centre for Nutrition and GI Diseases, Adelaide Medical School, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide 5000, Australia.
| |
Collapse
|
46
|
Cook BE, Membreno R, Zeglis BM. Dendrimer Scaffold for the Amplification of In Vivo Pretargeting Ligations. Bioconjug Chem 2018; 29:2734-2740. [PMID: 29969558 DOI: 10.1021/acs.bioconjchem.8b00385] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The development of immunoconjugates requires a careful balance between preserving the functionality of the antibody and modifying the immunoglobulin with the desired cargo. Herein, we describe the synthesis, development, and in vivo evaluation of a novel bifunctional dendrimeric scaffold and its application in pretargeted PET imaging. The site-specific modification of the huA33 antibody with this dendrimeric scaffold yields an immunoconjugate-sshuA33-DEN-TCO-decorated with ∼8 trans-cyclooctene (TCO) moieties, a marked increase compared to the ∼2 TCO/mAb of a nondendrimeric control immunoconjugate (sshuA33-PEG12-TCO). Pretargeted PET imaging and biodistribution experiments were used to compare the in vivo performance of these two immunoconjugates in athymic nude mice bearing subcutaneous SW1222 human colorectal cancer xenografts. To this end, the mice were administered 100 μg of each immunoconjugate followed 120 h later by the injection of a tetrazine-bearing radioligand, [64Cu]Cu-SarAr-Tz. Pretargeting with sshuA33-DEN-TCO produced excellent tumoral uptake at 24 h (8.9 ± 1.9 %ID/g), more than double that created by sshuA33-PEG12-TCO (4.1 ± 1.3 %ID/g). Critically-and somewhat surprisingly-the attachment of the G0.5 dendrimeric structures did not hamper the in vivo behavior of the immunoconjugate, suggesting that this versatile bifunctional scaffold may have applications beyond pretargeting.
Collapse
Affiliation(s)
- Brendon E Cook
- Department of Chemistry , Hunter College of the City University of New York , 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 , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Rosemery Membreno
- Department of Chemistry , Hunter College of the City University of New York , 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
| | - Brian M Zeglis
- Department of Chemistry , Hunter College of the City University of New York , 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 , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States.,Department of Radiology , Weill Cornell Medical College , New York , New York 10065 , United States
| |
Collapse
|
47
|
Carter LM, Poty S, Sharma SK, Lewis JS. Preclinical optimization of antibody-based radiopharmaceuticals for cancer imaging and radionuclide therapy-Model, vector, and radionuclide selection. J Labelled Comp Radiopharm 2018; 61:611-635. [PMID: 29412489 PMCID: PMC6081268 DOI: 10.1002/jlcr.3612] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/19/2017] [Accepted: 01/16/2018] [Indexed: 12/25/2022]
Abstract
Intact antibodies and their truncated counterparts (eg, Fab, scFv fragments) are generally exquisitely specific and selective vectors, enabling recognition of individual cancer-associated molecular phenotypes against a complex and dynamic biomolecular background. Complementary alignment of these advantages with unique properties of radionuclides is a defining paradigm in both radioimmunoimaging and radioimmunotherapy, which remain some of the most adept and promising tools for cancer diagnosis and treatment. This review discusses how translational potency can be maximized through rational selection of antibody-nuclide couples for radioimmunoimaging/therapy in preclinical models.
Collapse
Affiliation(s)
- Lukas M Carter
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sophie Poty
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sai Kiran Sharma
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Departments of Radiology and Pharmacology, Weill Cornell Medical College, New York, New York, USA
- Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
48
|
Lyu Z, Kang L, Buuh ZY, Jiang D, McGuth JC, Du J, Wissler HL, Cai W, Wang RE. A Switchable Site-Specific Antibody Conjugate. ACS Chem Biol 2018; 13:958-964. [PMID: 29461804 DOI: 10.1021/acschembio.8b00107] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Genetic incorporation of unnatural amino acids (UAAs) provides a unique approach to the synthesis of site-specific antibody conjugates that are homogeneous and better defined constructs than random conjugates. Yet, the yield varies for every antibody, and the process is costly and time-consuming. We have developed a switchable αGCN4-Fab conjugate that incorporates UAA p-acetylphenylalanine. The GCN4 peptide is used as a switch, and antibodies fused by GCN4 can direct the αGCN4-Fab conjugate to target different cancer cells for diagnosis, imaging, or therapeutic treatment. More importantly, this switchable conjugate demonstrated an impressive potential for pretargeted imaging in vivo. This approach illustrates the utility of an orthogonal switch as a general strategy to endow versatility to a single antibody conjugate, which should facilitate the application of UAA-based site-specific conjugates for a host of biomedical uses in the future.
Collapse
Affiliation(s)
- Zhigang Lyu
- Department of Chemistry, Temple University, 1901 N. 13th Street, Philadelphia, Pennsylvania 19122, United States
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, 100034, China
- Departments of Radiology and Medical Physics, University of Wisconsin—Madison, Madison, Wisconsin 53705, United States
| | - Zakey Yusuf Buuh
- Department of Chemistry, Temple University, 1901 N. 13th Street, Philadelphia, Pennsylvania 19122, United States
| | - Dawei Jiang
- Departments of Radiology and Medical Physics, University of Wisconsin—Madison, Madison, Wisconsin 53705, United States
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Jeffrey C. McGuth
- Department of Chemistry, Temple University, 1901 N. 13th Street, Philadelphia, Pennsylvania 19122, United States
| | - Juanjuan Du
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Haley L. Wissler
- Department of Chemistry, Temple University, 1901 N. 13th Street, Philadelphia, Pennsylvania 19122, United States
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin—Madison, Madison, Wisconsin 53705, United States
| | - Rongsheng E. Wang
- Department of Chemistry, Temple University, 1901 N. 13th Street, Philadelphia, Pennsylvania 19122, United States
| |
Collapse
|
49
|
Oliveira BL, Guo Z, Bernardes GJL. Inverse electron demand Diels-Alder reactions in chemical biology. Chem Soc Rev 2018; 46:4895-4950. [PMID: 28660957 DOI: 10.1039/c7cs00184c] [Citation(s) in RCA: 725] [Impact Index Per Article: 103.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The emerging inverse electron demand Diels-Alder (IEDDA) reaction stands out from other bioorthogonal reactions by virtue of its unmatchable kinetics, excellent orthogonality and biocompatibility. With the recent discovery of novel dienophiles and optimal tetrazine coupling partners, attention has now been turned to the use of IEDDA approaches in basic biology, imaging and therapeutics. Here we review this bioorthogonal reaction and its promising applications for live cell and animal studies. We first discuss the key factors that contribute to the fast IEDDA kinetics and describe the most recent advances in the synthesis of tetrazine and dienophile coupling partners. Both coupling partners have been incorporated into proteins for tracking and imaging by use of fluorogenic tetrazines that become strongly fluorescent upon reaction. Selected notable examples of such applications are presented. The exceptional fast kinetics of this catalyst-free reaction, even using low concentrations of coupling partners, make it amenable for in vivo radiolabelling using pretargeting methodologies, which are also discussed. Finally, IEDDA reactions have recently found use in bioorthogonal decaging to activate proteins or drugs in gain-of-function strategies. We conclude by showing applications of the IEDDA reaction in the construction of biomaterials that are used for drug delivery and multimodal imaging, among others. The use and utility of the IEDDA reaction is interdisciplinary and promises to revolutionize chemical biology, radiochemistry and materials science.
Collapse
Affiliation(s)
- B L Oliveira
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| | - Z Guo
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| | - G J L Bernardes
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK. and Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, Lisboa, 1649-028, Portugal.
| |
Collapse
|
50
|
Billaud EMF, Belderbos S, Cleeren F, Maes W, Van de Wouwer M, Koole M, Verbruggen A, Himmelreich U, Geukens N, Bormans G. Pretargeted PET Imaging Using a Bioorthogonal 18F-Labeled trans-Cyclooctene in an Ovarian Carcinoma Model. Bioconjug Chem 2017; 28:2915-2920. [PMID: 29191024 DOI: 10.1021/acs.bioconjchem.7b00635] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In cancer research, pretargeted positron emission tomography (PET) imaging has emerged as an effective two-step approach that combines the excellent target affinity and selectivity of antibodies with the advantages of using short-lived radionuclides such as fluorine-18. One possible approach is based on the bioorthogonal inverse-electron-demand Diels-Alder (IEDDA) reaction between tetrazines and trans-cyclooctene (TCO) derivatives. Here, we report the first successful use of an 18F-labeled small TCO compound, [18F]1 recently developed in our laboratory, to perform pretargeted immuno-PET imaging. The study was performed in an ovarian carcinoma mouse model, using a trastuzumab-tetrazine conjugate.
Collapse
Affiliation(s)
- Emilie M F Billaud
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven , Campus Gasthuisberg, O&N2, Herestraat 49, Box 821, 3000 Leuven, Belgium
| | - Sarah Belderbos
- Biomedical MRI/MoSAIC, Department of Imaging and Pathology, KU Leuven , Campus Gasthuisberg, O&N1, Herestraat 49, Box 505, 3000 Leuven, Belgium
| | - Frederik Cleeren
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven , Campus Gasthuisberg, O&N2, Herestraat 49, Box 821, 3000 Leuven, Belgium
| | - Wim Maes
- PharmAbs, the KU Leuven Antibody Center, KU Leuven , Campus Gasthuisberg, O&N2, Herestraat 49, Box 820, 3000 Leuven, Belgium
| | - Marlies Van de Wouwer
- PharmAbs, the KU Leuven Antibody Center, KU Leuven , Campus Gasthuisberg, O&N2, Herestraat 49, Box 820, 3000 Leuven, Belgium
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, University Hospital and KU Leuven , Herestraat 49, Box 7003, 3000 Leuven, Belgium
| | - Alfons Verbruggen
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven , Campus Gasthuisberg, O&N2, Herestraat 49, Box 821, 3000 Leuven, Belgium
| | - Uwe Himmelreich
- Biomedical MRI/MoSAIC, Department of Imaging and Pathology, KU Leuven , Campus Gasthuisberg, O&N1, Herestraat 49, Box 505, 3000 Leuven, Belgium
| | - Nick Geukens
- PharmAbs, the KU Leuven Antibody Center, KU Leuven , Campus Gasthuisberg, O&N2, Herestraat 49, Box 820, 3000 Leuven, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven , Campus Gasthuisberg, O&N2, Herestraat 49, Box 821, 3000 Leuven, Belgium
| |
Collapse
|