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
|
de Roode KE, Rossin R, Robillard MS. Toward Realization of Bioorthogonal Chemistry in the Clinic. Top Curr Chem (Cham) 2025; 383:12. [PMID: 40042792 PMCID: PMC11882664 DOI: 10.1007/s41061-025-00495-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025]
Abstract
In the last decade, the use of bioorthogonal chemistry toward medical applications has increased tremendously. Besides being useful for the production of pharmaceuticals, the efficient, nontoxic reactions open possibilities for the development of therapies that rely on in vivo chemistry between two bioorthogonal components. Here we discuss the latest developments in bioorthogonal chemistry, with a focus on their use in living organisms, the translation from model systems to humans, and the challenges encountered during preclinical development. We aim to provide the reader a broad presentation of the current state of the art and demonstrate the numerous possibilities that bioorthogonal reactions have for clinical use, now and in the near future.
Collapse
Affiliation(s)
- Kim E de Roode
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED, Nijmegen, The Netherlands
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Raffaella Rossin
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED, Nijmegen, The Netherlands
| | - Marc S Robillard
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED, Nijmegen, The Netherlands.
| |
Collapse
|
3
|
Börding T, Janik T, Bischoff P, Morkel M, Sers C, Horst D. GPA33 expression in colorectal cancer can be induced by WNT inhibition and targeted by cellular therapy. Oncogene 2025; 44:30-41. [PMID: 39472498 DOI: 10.1038/s41388-024-03200-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 01/07/2025]
Abstract
GPA33 is a promising surface antigen for targeted therapy in colorectal cancer (CRC). It is expressed almost exclusively in CRC and intestinal epithelia. However, previous clinical studies have not achieved expected response rates. We investigated GPA33 expression and regulation in CRC and developed a GPA33-targeted cellular therapy. We examined GPA33 expression in CRC cohorts using immunohistochemistry and immunofluorescence. We analyzed GPA33 regulation by interference with oncogenic signaling in vitro and in vivo using inhibitors and conditional inducible regulators. Furthermore, we engineered anti-GPA33-CAR T cells and assessed their activity in vitro and in vivo. GPA33 expression showed consistent intratumoral heterogeneity in CRC with antigen loss at the infiltrative tumor edge. This pattern was preserved at metastatic sites. GPA33-positive cells had a differentiated phenotype and low WNT activity. Low GPA33 expression levels were linked to tumor progression in patients with CRC. Downregulation of WNT activity induced GPA33 expression in vitro and in GPA33-negative tumor cell subpopulations in xenografts. GPA33-CAR T cells were activated in response to GPA33 and reduced xenograft growth in mice after intratumoral application. GPA33-targeted therapy may be improved by simultaneous WNT inhibition to enhance GPA33 expression. Furthermore, GPA33 is a promising target for cellular immunotherapy in CRC.
Collapse
Affiliation(s)
- Teresa Börding
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tobias Janik
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Philip Bischoff
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK) Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin, Germany
| | - Markus Morkel
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK) Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christine Sers
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK) Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David Horst
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- German Cancer Consortium (DKTK) Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
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
|
Rubahamya B, Dong S, Thurber GM. Clinical translation of antibody drug conjugate dosing in solid tumors from preclinical mouse data. SCIENCE ADVANCES 2024; 10:eadk1894. [PMID: 38820153 PMCID: PMC11141632 DOI: 10.1126/sciadv.adk1894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 04/29/2024] [Indexed: 06/02/2024]
Abstract
Antibody drug conjugates (ADCs) have made impressive strides in the clinic in recent years with 11 Food and Drug Administration approvals, including 6 for the treatment of patients with solid tumors. Despite this success, the development of new agents remains challenging with a high failure rate in the clinic. Here, we show that current approved ADCs for the treatment of patients with solid tumors can all show substantial efficacy in some mouse models when administered at a similar weight-based [milligrams per kilogram (mg/kg)] dosing in mice that is tolerated in the clinic. Mechanistically, equivalent mg/kg dosing results in a similar drug concentration in the tumor and a similar tissue penetration into the tumor due to the unique delivery features of ADCs. Combined with computational approaches, which can account for the complex distribution within the tumor microenvironment, these scaling concepts may aid in the evaluation of new agents and help design therapeutics with maximum clinical efficacy.
Collapse
Affiliation(s)
- Baron Rubahamya
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shujun Dong
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Greg M. Thurber
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
6
|
Sarrett S, Rodriguez C, Delaney S, Hosny MM, Sebastiano J, Santos-Coquillat A, Keinänen OM, Carter LM, Lastwika KJ, Lampe PD, Zeglis BM. Evaluating CD133 as a Radiotheranostic Target in Small-Cell Lung Cancer. Mol Pharm 2024; 21:1402-1413. [PMID: 38331430 PMCID: PMC10915790 DOI: 10.1021/acs.molpharmaceut.3c01063] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/19/2024] [Accepted: 01/19/2024] [Indexed: 02/10/2024]
Abstract
Despite decades of work, small-cell lung cancer (SCLC) remains a frustratingly recalcitrant disease. Both diagnosis and treatment are challenges: low-dose computed tomography (the approved method used for lung cancer screening) is unable to reliably detect early SCLC, and the malignancy's 5 year survival rate stands at a paltry 7%. Clearly, the development of novel diagnostic and therapeutic tools for SCLC is an urgent, unmet need. CD133 is a transmembrane protein that is expressed at low levels in normal tissue but is overexpressed by a variety of tumors, including SCLC. We previously explored CD133 as a biomarker for a novel autoantibody-to-immunopositron emission tomography (PET) strategy for the diagnosis of SCLC, work that first suggested the promise of the antigen as a radiotheranostic target in the disease. Herein, we report the in vivo validation of a pair of CD133-targeted radioimmunoconjugates for the PET imaging and radioimmunotherapy of SCLC. To this end, [89Zr]Zr-DFO-αCD133 was first interrogated in a trio of advanced murine models of SCLC─i.e., orthotopic, metastatic, and patient-derived xenografts─with the PET probe consistently producing high activity concentrations (>%ID/g) in tumor lesions combined with low uptake in healthy tissues. Subsequently, a variant of αCD133 labeled with the β-emitting radiometal 177Lu─[177Lu]Lu-DTPA-A″-CHX-αCD133─was synthesized and evaluated in a longitudinal therapy study in a subcutaneous xenograft model of SCLC, ultimately revealing that treatment with a dose of 9.6 MBq of the radioimmunoconjugate produced a significant increase in median survival compared to a control cohort. Taken together, these data establish CD133 as a viable target for the nuclear imaging and radiopharmaceutical therapy of SCLC.
Collapse
Affiliation(s)
- Samantha
M. Sarrett
- 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
| | - Cindy Rodriguez
- 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
| | - 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
| | - Meena M. Hosny
- Department
of Chemistry, Hunter College, City University
of New York, New York, New York 10065, United States
| | - Joni Sebastiano
- 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
| | - Ana Santos-Coquillat
- Department
of Chemistry, CICECO—Aveiro Institute of Materials, University of Aveiro, Campus Universitario de Santiago, Aveiro 3810-193, Portugal
| | - Outi M. Keinänen
- 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
- Department
of Chemistry, University of Helsinki, Helsinki 00100, Finland
| | - Lukas M. Carter
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
| | - Kristin J. Lastwika
- Translational
Research Program, Public Health Sciences
Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, United States
- Translational
Science and Therapeutics Division, Fred
Hutchinson Cancer Research Center, Seattle, Washington 98109, United States
| | - Paul D. Lampe
- Translational
Research Program, Public Health Sciences
Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, United States
- Human
Biology Division, Fred Hutchinson Cancer
Research Center, Seattle, Washington 98109, 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
| |
Collapse
|
7
|
Bauer D, Cornejo MA, Hoang TT, Lewis JS, Zeglis BM. Click Chemistry and Radiochemistry: An Update. Bioconjug Chem 2023; 34:1925-1950. [PMID: 37737084 PMCID: PMC10655046 DOI: 10.1021/acs.bioconjchem.3c00286] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/16/2023] [Indexed: 09/23/2023]
Abstract
The term "click chemistry" describes a class of organic transformations that were developed to make chemical synthesis simpler and easier, in essence allowing chemists to combine molecular subunits as if they were puzzle pieces. Over the last 25 years, the click chemistry toolbox has swelled from the canonical copper-catalyzed azide-alkyne cycloaddition to encompass an array of ligations, including bioorthogonal variants, such as the strain-promoted azide-alkyne cycloaddition and the inverse electron-demand Diels-Alder reaction. Without question, the rise of click chemistry has impacted all areas of chemical and biological science. Yet the unique traits of radiopharmaceutical chemistry have made it particularly fertile ground for this technology. In this update, we seek to provide a comprehensive guide to recent developments at the intersection of click chemistry and radiopharmaceutical chemistry and to illuminate several exciting trends in the field, including the use of emergent click transformations in radiosynthesis, the clinical translation of novel probes synthesized using click chemistry, and the advent of click-based in vivo pretargeting.
Collapse
Affiliation(s)
- David Bauer
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10021, United States
| | - Mike A. Cornejo
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10021, United States
- Department
of Chemistry, Hunter College, City University
of New York, New York, New York 10065, United States
- Ph.D.
Program in Chemistry, Graduate Center of
the City University of New York, New York, New York 10016, United States
| | - Tran T. Hoang
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10021, United States
- Department
of Pharmacology, Weill Cornell Medical College, New York, New York 10065, United States
| | - Jason S. Lewis
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10021, United States
- Department
of Radiology, Weill Cornell Medical College, New York 10021, New York United States
| | - Brian M. Zeglis
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10021, United States
- Department
of Chemistry, Hunter College, City University
of New York, New York, New York 10065, United States
- Ph.D.
Program in Chemistry, Graduate Center of
the City University of New York, New York, New York 10016, United States
- Department
of Pharmacology, Weill Cornell Medical College, New York, New York 10065, United States
- Department
of Radiology, Weill Cornell Medical College, New York 10021, New York United States
- Ph.D.
Program
in Biochemistry, Graduate Center of the
City University of New York, New
York, New York 10016, United States
| |
Collapse
|
8
|
Wu Z, Dou J, Nguyen KU, Eppley JC, Siwawannapong K, Zhang Y, Lindsey JS. Tailoring the AIE Chromogen 2-(2-Hydroxyphenyl)benzothiazole for Use in Enzyme-Triggered Molecular Brachytherapy. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248682. [PMID: 36557815 PMCID: PMC9786593 DOI: 10.3390/molecules27248682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022]
Abstract
A targeted strategy for treating cancer is antibody-directed enzyme prodrug therapy, where the enzyme attached to the antibody causes conversion of an inactive small-molecule prodrug into an active drug. A limitation may be the diffusion of the active drug away from the antibody target site. A related strategy with radiotherapeutics entails enzymatically promoted conversion of a soluble to insoluble radiotherapeutic agent, thereby immobilizing the latter at the target site. Such a molecular brachytherapy has been scarcely investigated. In distinct research, the advent of molecular designs for aggregation-induced emission (AIE) suggests translational use in molecular brachytherapy. Here, several 2-(2-hydroxyphenyl)benzothiazole substrates that readily aggregate in aqueous solution (and afford AIE) were elaborated in this regard. In particular, (1) the 2-(2-hydroxyphenyl) unit was derivatized to bear a pegylated phosphodiester that imparts water solubility yet undergoes enzymatic cleavage, and (2) a p-phenol unit was attached to the benzo moiety to provide a reactive site for final-step iodination (here examined with natural abundance iodide). The pegylated phosphodiester-iodinated benzothiazole undergoes conversion from aqueous-soluble to aqueous-insoluble upon treatment with a phosphatase or phosphodiesterase. The aggregation is essential to molecular brachytherapy, whereas the induced emission of AIE is not essential but provides a convenient basis for research development. Altogether, 21 compounds were synthesized (18 new, 3 known via new routes). Taken together, blending biomedical strategies of enzyme prodrug therapy with materials chemistry concerning substances that undergo AIE may comprise a step forward on the long road toward molecular brachytherapy.
Collapse
|
9
|
Cheal SM, Chung SK, Vaughn BA, Cheung NKV, Larson SM. Pretargeting: A Path Forward for Radioimmunotherapy. J Nucl Med 2022; 63:1302-1315. [PMID: 36215514 PMCID: PMC12079710 DOI: 10.2967/jnumed.121.262186] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/07/2022] [Indexed: 12/19/2022] Open
Abstract
Pretargeted radioimmunodiagnosis and radioimmunotherapy aim to efficiently combine antitumor antibodies and medicinal radioisotopes for high-contrast imaging and high-therapeutic-index (TI) tumor targeting, respectively. As opposed to conventional radioimmunoconjugates, pretargeted approaches separate the tumor-targeting step from the payload step, thereby amplifying tumor uptake while reducing normal-tissue exposure. Alongside contrast and TI, critical parameters include antibody immunogenicity and specificity, availability of radioisotopes, and ease of use in the clinic. Each of the steps can be optimized separately; as modular systems, they can find broad applications irrespective of tumor target, tumor type, or radioisotopes. Although this versatility presents enormous opportunity, pretargeting is complex and presents unique challenges for clinical translation and optimal use in patients. The purpose of this article is to provide a brief historical perspective on the origins and development of pretargeting strategies in nuclear medicine, emphasizing 2 protein delivery systems that have been extensively evaluated (i.e., biotin-streptavidin and hapten-bispecific monoclonal antibodies), as well as radiohaptens and radioisotopes. We also highlight recent innovations, including pretargeting with bioorthogonal chemistry and novel protein vectors (such as self-assembling and disassembling proteins and Affibody molecules). We caution the reader that this is by no means a comprehensive review of the past 3 decades of pretargeted radioimmunodiagnosis and pretargeted radioimmunotherapy. But we do aim to highlight major developmental milestones and to identify benchmarks for success with regard to TI and toxicity in preclinical models and clinically. We believe this approach will lead to the identification of key obstacles to clinical success, revive interest in the utility of radiotheranostics applications, and guide development of the next generation of pretargeted theranostics.
Collapse
Affiliation(s)
- Sarah M Cheal
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York;
| | - Sebastian K Chung
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brett A Vaughn
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Steven M Larson
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
10
|
Jallinoja VIJ, Carney BD, Bhatt K, Abbriano CH, Schlyer DJ, Yazaki PJ, Houghton JL. Investigation of Copper-64-Based Host-Guest Chemistry Pretargeted Positron Emission Tomography. Mol Pharm 2022; 19:2268-2278. [PMID: 35700402 PMCID: PMC11271262 DOI: 10.1021/acs.molpharmaceut.2c00102] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Pretargeting is a technique that uses macromolecules as targeting agents for nuclear imaging and therapy with the goal of reducing the radiation toxicity to healthy tissues often associated with directly radiolabeled macromolecules. In pretargeting, a macromolecule is radiolabeled in vivo at the target site using a radiolabeled small molecule (radioligand) that interacts with the macromolecule with high specificity. We report an investigation of host-guest chemistry-driven pretargeting using copper-64 radiolabeled ferrocene (Fc; guest) compounds and a cucurbit[7]uril (CB7; host) molecule functionalized carcinoembryonic antigen targeting hT84.66-M5A monoclonal antibody (CB7-M5A). Two novel ferrocene-based radioligands ([64Cu]Cu-NOTA-PEG3-Fc and [64Cu]Cu-NOTA-PEG7-Fc) were prepared, and their in vitro stability, pharmacokinetic in vivo profile in healthy mice, and pretargeting performance in a subcutaneous BxPC3 human pancreatic cancer cell xenograft mouse model were compared. The antibody dosing was optimized using a zirconium-89 radiolabeled M5A antibody ([89Zr]Zr-DFO-M5A) in a BxPC3 xenograft model, and the dosimetry of [89Zr]Zr-DFO-M5A and the pretargeting approach were compared. Finally, the effects of varying lag times up to 9 days between CB7-M5A and radioligand injection were investigated. In vivo pretargeting studies with both ferrocene radioligands resulted in specific tumor uptake (p = 0.0006 and p = 0.003) and also showed that the host-guest-based pretargeting approach excels with extended lag times up to 9 days with good tumor localization, suggesting that host-guest pretargeting may be suitable for use without clearing agents which have complicated clinical application of this technique. To our knowledge, the reported lag time of 9 days is the longest investigated lag time in any reported pretargeting studies.
Collapse
Affiliation(s)
- Vilma I J Jallinoja
- Department of Radiology, Stony Brook University, Stony Brook, New York 11794, United States
- Chemical and Physical Biology Graduate Program, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Brandon D Carney
- Department of Radiology, Stony Brook University, Stony Brook, New York 11794, United States
| | - Kavita Bhatt
- Department of Radiology, Stony Brook University, Stony Brook, New York 11794, United States
| | - Courtney H Abbriano
- Department of Radiology, Stony Brook University, Stony Brook, New York 11794, United States
| | - David J Schlyer
- Department of Radiology, Stony Brook University, Stony Brook, New York 11794, United States
- Collider-Accelerator Department, Brookhaven National Laboratory, Upton, New York 11973, United States
| | - Paul J Yazaki
- Beckman Institute, City of Hope, Duarte, California 91010, United States
| | - Jacob L Houghton
- Department of Radiology, Stony Brook University, Stony Brook, New York 11794, United States
| |
Collapse
|
11
|
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
|
12
|
Pan Y, Tang W, Fan W, Zhang J, Chen X. Development of nanotechnology-mediated precision radiotherapy for anti-metastasis and radioprotection. Chem Soc Rev 2022; 51:9759-9830. [DOI: 10.1039/d1cs01145f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Radiotherapy (RT), including external beam RT and internal radiation therapy, uses high-energy ionizing radiation to kill tumor cells.
Collapse
Affiliation(s)
- Yuanbo Pan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
| | - Wei Tang
- Departments of Pharmacy and Diagnostic Radiology, Nanomedicine Translational Research Program, Faculty of Science and Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117544, Singapore
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Sarrett SM, Keinänen O, Dayts EJ, Dewaele-Le Roi G, Rodriguez C, Carnazza KE, Zeglis BM. Inverse electron demand Diels-Alder click chemistry for pretargeted PET imaging and radioimmunotherapy. Nat Protoc 2021; 16:3348-3381. [PMID: 34127865 PMCID: PMC8917728 DOI: 10.1038/s41596-021-00540-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 03/22/2021] [Indexed: 11/08/2022]
Abstract
Radiolabeled antibodies have shown promise as tools for both the nuclear imaging and endoradiotherapy of cancer, but the protracted circulation time of radioimmunoconjugates can lead to high radiation doses to healthy tissues. To circumvent this issue, we have developed an approach to positron emission tomography (PET) imaging and radioimmunotherapy (RIT) predicated on radiolabeling the antibody after it has reached its target within the body. This in vivo pretargeting strategy is based on the rapid and bio-orthogonal inverse electron demand Diels-Alder reaction between tetrazine (Tz) and trans-cyclooctene (TCO). Pretargeted PET imaging and RIT using TCO-modified antibodies in conjunction with Tz-bearing radioligands produce high activity concentrations in target tissues as well as reduced radiation doses to healthy organs compared to directly labeled radioimmunoconjugates. Herein, we describe how to prepare a TCO-modified antibody (humanized A33-TCO) as well as how to synthesize two Tz-bearing radioligands: one labeled with the positron-emitting radiometal copper-64 ([64Cu]Cu-SarAr-Tz) and one labeled with the β-emitting radiolanthanide lutetium-177 ([177Lu]Lu-DOTA-PEG7-Tz). We also provide a detailed description of pretargeted PET and pretargeted RIT experiments in a murine model of human colorectal carcinoma. Proper training in both radiation safety and the handling of laboratory mice is required for the successful execution of this protocol.
Collapse
Affiliation(s)
- Samantha M Sarrett
- Department of Chemistry, Hunter College, City University of New York, New York, NY, USA
- PhD Program in Biochemistry, Graduate Center of the City University of New York, New York, NY, USA
| | - Outi Keinänen
- Department of Chemistry, Hunter College, City University of New York, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Chemistry, Radiochemistry, University of Helsinki, Helsinki, Finland
| | - Eric J Dayts
- Department of Chemistry, Hunter College, City University of New York, New York, NY, USA
| | - Guillaume Dewaele-Le Roi
- Department of Chemistry, Hunter College, 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, Graduate Center of the City University of New York, New York, NY, USA
| | - Cindy Rodriguez
- Department of Chemistry, Hunter College, City University of New York, New York, NY, USA
- Ph.D. Program in Chemistry, Graduate Center of the City University of New York, New York, NY, USA
| | - Kathryn E Carnazza
- Brain and Mind Research Institute & Appel Institute for Alzheimer's Disease Research, Weill Cornell Medical College, New York, NY, USA
| | - Brian M Zeglis
- Department of Chemistry, Hunter College, City University of New York, New York, NY, USA.
- PhD Program in Biochemistry, 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, 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
|
16
|
Guillou A, Earley DF, Klingler S, Nisli E, Nüesch LJ, Fay R, Holland JP. The Influence of a Polyethylene Glycol Linker on the Metabolism and Pharmacokinetics of a 89Zr-Radiolabeled Antibody. Bioconjug Chem 2021; 32:1263-1275. [PMID: 34056896 DOI: 10.1021/acs.bioconjchem.1c00172] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Most experimental work in the space of bioconjugation chemistry focuses on using new methods to construct covalent bonds between a cargo molecule and a protein of interest such as a monoclonal antibody (mAb). Bond formation is important for generating new diagnostic tools, yet when these compounds advance to preclinical in vitro and in vivo studies, and later for translation to the clinic, understanding the fate of potential metabolites that arise from chemical or enzymatic degradation of the construct is important to obtain a full picture of the pharmacokinetic performance of a new compound. In the context of designing new bioconjugate methods for labeling antibodies with the positron-emitting radionuclide 89Zr, we previously developed a photochemical process for making 89Zr-mAbs. Experimental studies on [89Zr]ZrDFO-PEG3-azepin-mAb constructs revealed that incorporation of the tris-polyethylene glycol (PEG3) linker improved the aqueous phase solubility and radiochemical conversion. However, the use of a PEG3 linker also has an impact on the whole-body residence time of the construct, leading to a more rapid excretion of the 89Zr activity when compared with radiotracers that lack the PEG3 chain. In this work, we investigated the metabolic fate of eight possible metabolites that arise from the logical disconnection of [89Zr]ZrDFO-PEG3-azepin-mAb at bonds which are susceptible to chemical or enzymatic cleavage. Synthesis combined with 89Zr-radiolabeling, small-animal positron emission tomography imaging at multiple time points from 0 to 20 h, and measurements of the effective half-life for whole-body excretion are reported. The conclusions are that the use of a PEG3 linker is non-innocent in terms of its impact on enhancing the metabolism of [89Zr]ZrDFO-PEG3-azepin-mAbs. In most cases, degradation can produce metabolites that are rapidly eliminated from the body, thereby enhancing image contrast by reducing nonspecific accumulation and retention of 89Zr in background organs such as the liver, spleen, kidney, and bone.
Collapse
Affiliation(s)
- Amaury Guillou
- Department of Chemistry University of Zurich Winterthurerstrasse 190 CH-8057, Zurich, Switzerland
| | - Daniel F Earley
- Department of Chemistry University of Zurich Winterthurerstrasse 190 CH-8057, Zurich, Switzerland
| | - Simon Klingler
- Department of Chemistry University of Zurich Winterthurerstrasse 190 CH-8057, Zurich, Switzerland
| | - Eda Nisli
- Department of Chemistry University of Zurich Winterthurerstrasse 190 CH-8057, Zurich, Switzerland
| | - Laura J Nüesch
- Department of Chemistry University of Zurich Winterthurerstrasse 190 CH-8057, Zurich, Switzerland
| | - Rachael Fay
- Department of Chemistry University of Zurich Winterthurerstrasse 190 CH-8057, Zurich, Switzerland
| | - Jason P Holland
- Department of Chemistry University of Zurich Winterthurerstrasse 190 CH-8057, Zurich, Switzerland
| |
Collapse
|
17
|
White JM, Escorcia FE, Viola NT. Perspectives on metals-based radioimmunotherapy (RIT): moving forward. Theranostics 2021; 11:6293-6314. [PMID: 33995659 PMCID: PMC8120204 DOI: 10.7150/thno.57177] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/22/2021] [Indexed: 12/18/2022] Open
Abstract
Radioimmunotherapy (RIT) is FDA-approved for the clinical management of liquid malignancies, however, its use for solid malignancies remains a challenge. The putative benefit of RIT lies in selective targeting of antigens expressed on the tumor surface using monoclonal antibodies, to systemically deliver cytotoxic radionuclides. The past several decades yielded dramatic improvements in the quality, quantity, recent commercial availability of alpha-, beta- and Auger Electron-emitting therapeutic radiometals. Investigators have created new or improved existing bifunctional chelators. These bifunctional chelators bind radiometals and can be coupled to antigen-specific antibodies. In this review, we discuss approaches to develop radiometal-based RITs, including the selection of radiometals, chelators and antibody platforms (i.e. full-length, F(ab')2, Fab, minibodies, diabodies, scFv-Fc and nanobodies). We cite examples of the performance of RIT in the clinic, describe challenges to its implementation, and offer insights to address gaps toward translation.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/therapeutic use
- Antigens, Neoplasm/immunology
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/metabolism
- Antineoplastic Agents, Immunological/therapeutic use
- Chelating Agents/administration & dosage
- Chelating Agents/metabolism
- Click Chemistry
- Clinical Trials as Topic
- Dose Fractionation, Radiation
- Drug Delivery Systems
- Forecasting
- Humans
- Immunoglobulin Fab Fragments/administration & dosage
- Immunoglobulin Fab Fragments/therapeutic use
- Lymphoma, Non-Hodgkin/radiotherapy
- Mice
- Molecular Targeted Therapy
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasms, Experimental/diagnostic imaging
- Neoplasms, Experimental/radiotherapy
- Organ Specificity
- Precision Medicine
- Radiation Tolerance
- Radioimmunotherapy/methods
- Radiopharmaceuticals/administration & dosage
- Radiopharmaceuticals/therapeutic use
- Receptor Protein-Tyrosine Kinases/antagonists & inhibitors
- Single-Chain Antibodies/administration & dosage
- Single-Chain Antibodies/therapeutic use
- Single-Domain Antibodies/administration & dosage
- Single-Domain Antibodies/therapeutic use
- Yttrium Radioisotopes/administration & dosage
- Yttrium Radioisotopes/therapeutic use
Collapse
Affiliation(s)
- Jordan M. White
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201
- Department of Oncology, Karmanos Cancer Institute, Detroit, MI 48201
| | - Freddy E. Escorcia
- Molecular Imaging Branch, Radiation Oncology Branch, National Cancer Institute, Bethesda, MD 20814
| | - Nerissa T. Viola
- Department of Oncology, Karmanos Cancer Institute, Detroit, MI 48201
| |
Collapse
|
18
|
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]
|
19
|
Qiu L, Lin Q, Si Z, Tan H, Liu G, Zhou J, Wang T, Chen Y, Huang Y, Yu T, Jin M, Cheng D, Shi H. A Pretargeted Imaging Strategy for EGFR-Positive Colorectal Carcinoma via Modulation of Tz-Radioligand Pharmacokinetics. Mol Imaging Biol 2021; 23:38-51. [PMID: 32914391 DOI: 10.1007/s11307-020-01539-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 11/27/2022]
Abstract
PURPOSE Previously, we successfully developed a pretargeted imaging strategy (atezolizumab-TCO/[99mTc]HYNIC-PEG11-Tz) for evaluating programmed cell death ligand-1 (PD-L1) expression in xenograft mice. However, the surplus unclicked [99mTc]HYNIC-PEG11-Tz is cleared somewhat sluggishly through the intestines, which is not ideal for colorectal cancer (CRC) imaging. To shift the excretion of the Tz-radioligand to the renal system, we developed a novel Tz-radioligand by adding a polypeptide linker between HYNIC and PEG11. PROCEDURES Pretargeted molecular probes [99mTc]HYNIC-polypeptide-PEG11-Tz and cetuximab-TCO were synthesized. [99mTc]HYNIC-polypeptide-PEG11-Tz was evaluated for in vitro stability and in vivo blood pharmacokinetics. In vitro ligation reactivity of [99mTc]HYNIC-polypeptide-PEG11-Tz towards cetuximab-TCO was also tested. Biodistribution assay and imaging of [99mTc]HYNIC-polypeptide-PEG11-Tz were performed to observe its excretion pathway. Pretargeted biodistribution was measured at three different accumulation intervals to determine the optimal pretargeted interval time. Pretargeted (cetuximab-TCO 48 h/[99mTc]HYNIC-PEG11-Tz 6 h) and (cetuximab-TCO 48 h/[99mTc]HYNIC-Polypeptide-PEG11-Tz 6 h) imagings were compared to examine the effect of the excretion pathway on tumor imaging. RESULTS [99mTc]HYNIC-polypeptide-PEG11-Tz showed favorable in vitro stability and rapid blood clearance in mice. SEC-HPLC revealed almost complete reaction between cetuximab-TCO and [99mTc]HYNIC-polypeptide-PEG11-Tz in vitro, with the 8:1 Tz-to-mAb reaction providing a conversion yield of 87.83 ± 3.27 %. Biodistribution and imaging analyses showed that the Tz-radioligand was cleared through the kidneys. After 24, 48, and 72 h of accumulation in HCT116 tumor, the tumor-to-blood ratio of cetuximab-TCO was 0.83 ± 0.13, 1.40 ± 0.31, and 1.15 ± 0.21, respectively. Both pretargeted (cetuximab-TCO 48 h/[99mTc]HYNIC-PEG11-Tz 6 h) and (cetuximab-TCO 48 h/[99mTc]HYNIC-polypeptide-PEG11-Tz 6 h) clearly delineated HCT116 tumor. Pretargeted imaging strategy using cetuximab-TCO/[99mTc]HYNIC-polypeptide-PEG11-Tz could be used for diagnosing CRC, as the surplus unclicked [99mTc]HYNIC-polypeptide-PEG11-Tz was cleared through the urinary system, leading to low abdominal uptake background. CONCLUSION Our novel pretargeted imaging strategy (cetuximab-TCO/[99mTc]HYNIC-polypeptide-PEG11-Tz) was useful for imaging CRC, broadening the application scope of pretargeted imaging strategy. The pretargeted imaging strategy clearly delineated HCT116 tumor, showing that its use could be extended to selection of internalizing antibodies.
Collapse
Affiliation(s)
- Lin Qiu
- Department of Nuclear Medicine, Zhongshan Hospital Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, China
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Qingyu Lin
- Department of Nuclear Medicine, Zhongshan Hospital Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Zhan Si
- Department of Nuclear Medicine, Zhongshan Hospital Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Hui Tan
- Department of Nuclear Medicine, Zhongshan Hospital Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Guobing Liu
- Department of Nuclear Medicine, Zhongshan Hospital Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Jun Zhou
- Department of Nuclear Medicine, Zhongshan Hospital Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Tingting Wang
- Department of Nuclear Medicine, Zhongshan Hospital Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Yue Chen
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | | | - Tao Yu
- WuXi AppTec, Shanghai, China
| | - Mingzhi Jin
- WuXi Biologics (Shanghai) Co., Ltd, Shanghai, China
| | - Dengfeng Cheng
- Department of Nuclear Medicine, Zhongshan Hospital Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, China.
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, China.
| |
Collapse
|
20
|
Nicolson F, Kircher MF. Theranostics: Agents for Diagnosis and Therapy. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00040-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
21
|
Porte K, Riberaud M, Châtre R, Audisio D, Papot S, Taran F. Bioorthogonal Reactions in Animals. Chembiochem 2020; 22:100-113. [PMID: 32935888 DOI: 10.1002/cbic.202000525] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/15/2020] [Indexed: 01/04/2023]
Abstract
The advent of bioorthogonal chemistry has led to the development of powerful chemical tools that enable increasingly ambitious applications. In particular, these tools have made it possible to achieve what is considered to be the holy grail of many researchers involved in chemical biology: to perform unnatural chemical reactions within living organisms. In this minireview, we present an update of bioorthogonal reactions that have been carried out in animals for various applications. We outline the advances made in the understanding of fundamental biological processes, and the development of innovative imaging and therapeutic strategies using bioorthogonal chemistry.
Collapse
Affiliation(s)
- Karine Porte
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191, Gif-sur-Yvette, France
| | - Maxime Riberaud
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191, Gif-sur-Yvette, France
| | - Rémi Châtre
- Université de Poitiers, UMR-CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), 86022, Poitiers, France) E-mail
| | - Davide Audisio
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191, Gif-sur-Yvette, France
| | - Sébastien Papot
- Université de Poitiers, UMR-CNRS 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), 86022, Poitiers, France) E-mail
| | - Frédéric Taran
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191, Gif-sur-Yvette, France
| |
Collapse
|
22
|
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
|
23
|
Béquignat JB, Ty N, Rondon A, Taiariol L, Degoul F, Canitrot D, Quintana M, Navarro-Teulon I, Miot-Noirault E, Boucheix C, Chezal JM, Moreau E. Optimization of IEDDA bioorthogonal system: Efficient process to improve trans-cyclooctene/tetrazine interaction. Eur J Med Chem 2020; 203:112574. [PMID: 32683167 DOI: 10.1016/j.ejmech.2020.112574] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/11/2020] [Accepted: 06/11/2020] [Indexed: 10/23/2022]
Abstract
The antibody pretargeting approach for radioimmunotherapy (RIT) using inverse electron demand Diels-Alder cycloaddition (IEDDA) constitutes an emerging theranostic approach for solid cancers. However, IEDDA pretargeting has not reached clinical trial. The major limitation of the IEDDA strategy depends largely on trans-cyclooctene (TCO) stability. Indeed, TCO may isomerize into the more stable but unreactive cis-cyclooctene (CCO), leading to a drastic decrease of IEDDA efficiency. We have thus developed both efficient and reproducible synthetic pathways and analytical follow up for (PEGylated) TCO derivatives, providing high TCO isomeric purity for antibody modification. We have set up an original process to limit the isomerization of TCO to CCO before the mAbs' functionalization to allow high TCO/tetrazine cycloaddition.
Collapse
Affiliation(s)
- Jean-Baptiste Béquignat
- Université Clermont Auvergne, Imagerie Moléculaire et Stratégies Théranostiques, BP 184, F-63005, Clermont-Ferrand, France; Inserm, U 1240, F-63000, Clermont-Ferrand, France; Centre Jean Perrin, F-63011, Clermont-Ferrand, France
| | - Nancy Ty
- Université Clermont Auvergne, Imagerie Moléculaire et Stratégies Théranostiques, BP 184, F-63005, Clermont-Ferrand, France; Inserm, U 1240, F-63000, Clermont-Ferrand, France; Centre Jean Perrin, F-63011, Clermont-Ferrand, France
| | - Aurélie Rondon
- Université Clermont Auvergne, Imagerie Moléculaire et Stratégies Théranostiques, BP 184, F-63005, Clermont-Ferrand, France; Inserm, U 1240, F-63000, Clermont-Ferrand, France; Centre Jean Perrin, F-63011, Clermont-Ferrand, France; Institut de Recherche en Cancérologie (IRCM), U1194 - Université Montpellier - ICM, Radiobiology and Targeted Radiotherapy, 34298, Montpellier Cedex 5, France
| | - Ludivine Taiariol
- Université Clermont Auvergne, Imagerie Moléculaire et Stratégies Théranostiques, BP 184, F-63005, Clermont-Ferrand, France; Inserm, U 1240, F-63000, Clermont-Ferrand, France; Centre Jean Perrin, F-63011, Clermont-Ferrand, France
| | - Françoise Degoul
- Université Clermont Auvergne, Imagerie Moléculaire et Stratégies Théranostiques, BP 184, F-63005, Clermont-Ferrand, France; Inserm, U 1240, F-63000, Clermont-Ferrand, France; Centre Jean Perrin, F-63011, Clermont-Ferrand, France
| | - Damien Canitrot
- Université Clermont Auvergne, Imagerie Moléculaire et Stratégies Théranostiques, BP 184, F-63005, Clermont-Ferrand, France; Inserm, U 1240, F-63000, Clermont-Ferrand, France; Centre Jean Perrin, F-63011, Clermont-Ferrand, France
| | - Mercedes Quintana
- Université Clermont Auvergne, Imagerie Moléculaire et Stratégies Théranostiques, BP 184, F-63005, Clermont-Ferrand, France; Inserm, U 1240, F-63000, Clermont-Ferrand, France; Centre Jean Perrin, F-63011, Clermont-Ferrand, France
| | - Isabelle Navarro-Teulon
- Institut de Recherche en Cancérologie (IRCM), U1194 - Université Montpellier - ICM, Radiobiology and Targeted Radiotherapy, 34298, Montpellier Cedex 5, France
| | - Elisabeth Miot-Noirault
- Université Clermont Auvergne, Imagerie Moléculaire et Stratégies Théranostiques, BP 184, F-63005, Clermont-Ferrand, France; Inserm, U 1240, F-63000, Clermont-Ferrand, France; Centre Jean Perrin, F-63011, Clermont-Ferrand, France
| | | | - Jean-Michel Chezal
- Université Clermont Auvergne, Imagerie Moléculaire et Stratégies Théranostiques, BP 184, F-63005, Clermont-Ferrand, France; Inserm, U 1240, F-63000, Clermont-Ferrand, France; Centre Jean Perrin, F-63011, Clermont-Ferrand, France
| | - Emmanuel Moreau
- Université Clermont Auvergne, Imagerie Moléculaire et Stratégies Théranostiques, BP 184, F-63005, Clermont-Ferrand, France; Inserm, U 1240, F-63000, Clermont-Ferrand, France; Centre Jean Perrin, F-63011, Clermont-Ferrand, France.
| |
Collapse
|
24
|
Evaluation of Organo [ 18F]Fluorosilicon Tetrazine as a Prosthetic Group for the Synthesis of PET Radiotracers. Molecules 2020; 25:molecules25051208. [PMID: 32156020 PMCID: PMC7179430 DOI: 10.3390/molecules25051208] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 01/09/2023] Open
Abstract
Fluorine-18 is the most widely used positron emission tomography (PET) radionuclide currently in clinical application, due to its optimal nuclear properties. The synthesis of 18F-labeled radiotracers often requires harsh reaction conditions, limiting the use of sensitive bio- and macromolecules as precursors for direct radiolabeling with fluorine-18. We aimed to develop a milder and efficient in vitro and in vivo labeling method for trans-cyclooctene (TCO) functionalized proteins, through the bioorthogonal inverse-electron demand Diels-Alder (IEDDA) reaction with fluorine-18 radiolabeled tetrazine ([18F]SiFA-Tz). Here, we used TCO-modified bovine serum albumin (BSA) as the model protein, and isotopic exchange (IE) (19F/18F) chemistry as the labeling strategy. The radiolabeling of albumin-TCO with [18F]SiFA-Tz ([18F]6), providing [18F]fluoroalbumin ([18F]10) in high radiochemical yield (99.1 ± 0.2%, n = 3) and a molar activity (MA) of 1.1 GBq/µmol, confirmed the applicability of [18F]6 as a quick in vitro fluorination reagent for the TCO functionalized proteins. While the biological evaluation of [18F]6 demonstrated defluorination in vivo, limiting the utility for pretargeted applications, the in vivo stability of the radiotracer was dramatically improved when [18F]6 was used for the radiolabeling of albumin-TCO ([18F]10) in vitro, prior to administration. Due to the detected defluorination in vivo, structural optimization of the prosthetic group for improved stability is needed before further biological studies and application of pretargeted PET imaging.
Collapse
|
25
|
Ediriweera GR, Simpson JD, Fuchs AV, Venkatachalam TK, Van De Walle M, Howard CB, Mahler SM, Blinco JP, Fletcher NL, Houston ZH, Bell CA, Thurecht KJ. Targeted and modular architectural polymers employing bioorthogonal chemistry for quantitative therapeutic delivery. Chem Sci 2020; 11:3268-3280. [PMID: 34122834 PMCID: PMC8157365 DOI: 10.1039/d0sc00078g] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
There remain several key challenges to existing therapeutic systems for cancer therapy, such as quantitatively determining the true, tissue-specific drug release profile in vivo, as well as reducing side-effects for an increased standard of care. Hence, it is crucial to engineer new materials that allow for a better understanding of the in vivo pharmacokinetic/pharmacodynamic behaviours of therapeutics. We have expanded on recent “click-to-release” bioorthogonal pro-drug activation of antibody-drug conjugates (ADCs) to develop a modular and controlled theranostic system for quantitatively assessing site-specific drug activation and deposition from a nanocarrier molecule, by employing defined chemistries. The exploitation of quantitative imaging using positron emission tomography (PET) together with pre-targeted bioorthogonal chemistries in our system provided an effective means to assess in real-time the exact amount of active drug administered at precise sites in the animal; our methodology introduces flexibility in both the targeting and therapeutic components that is specific to nanomedicines and offers unique advantages over other technologies. In this approach, the in vivo click reaction facilitates pro-drug activation as well as provides a quantitative means to investigate the dynamic behaviour of the therapeutic agent. There remain several key challenges to existing therapeutic systems for cancer therapy, such as quantitatively determining the true, tissue-specific drug release profile in vivo, as well as reducing side-effects for an increased standard of care.![]()
Collapse
Affiliation(s)
- Gayathri R Ediriweera
- Centre for Advanced Imaging, The University of Queensland Brisbane QLD 4072 Australia .,Australian Institute for Bioengineering & Nanotechnology (AIBN), The University of Queensland Brisbane QLD 4072 Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland Brisbane QLD 4072 Australia
| | - Joshua D Simpson
- Centre for Advanced Imaging, The University of Queensland Brisbane QLD 4072 Australia .,Australian Institute for Bioengineering & Nanotechnology (AIBN), The University of Queensland Brisbane QLD 4072 Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland Brisbane QLD 4072 Australia
| | - Adrian V Fuchs
- Centre for Advanced Imaging, The University of Queensland Brisbane QLD 4072 Australia .,Australian Institute for Bioengineering & Nanotechnology (AIBN), The University of Queensland Brisbane QLD 4072 Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland Brisbane QLD 4072 Australia
| | - Taracad K Venkatachalam
- Centre for Advanced Imaging, The University of Queensland Brisbane QLD 4072 Australia .,Australian Institute for Bioengineering & Nanotechnology (AIBN), The University of Queensland Brisbane QLD 4072 Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland Brisbane QLD 4072 Australia
| | - Matthias Van De Walle
- School of Chemistry, Physics and Mechanical Engineering, Queensland University of Technology 2 George St Brisbane QLD 4000 Australia
| | - Christopher B Howard
- Australian Institute for Bioengineering & Nanotechnology (AIBN), The University of Queensland Brisbane QLD 4072 Australia.,ARC Training Centre for Biopharmaceutical Innovation, The University of Queensland Brisbane QLD 4072 Australia
| | - Stephen M Mahler
- Australian Institute for Bioengineering & Nanotechnology (AIBN), The University of Queensland Brisbane QLD 4072 Australia.,ARC Training Centre for Biopharmaceutical Innovation, The University of Queensland Brisbane QLD 4072 Australia
| | - James P Blinco
- School of Chemistry, Physics and Mechanical Engineering, Queensland University of Technology 2 George St Brisbane QLD 4000 Australia
| | - Nicholas L Fletcher
- Centre for Advanced Imaging, The University of Queensland Brisbane QLD 4072 Australia .,Australian Institute for Bioengineering & Nanotechnology (AIBN), The University of Queensland Brisbane QLD 4072 Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland Brisbane QLD 4072 Australia
| | - Zachary H Houston
- Centre for Advanced Imaging, The University of Queensland Brisbane QLD 4072 Australia .,Australian Institute for Bioengineering & Nanotechnology (AIBN), The University of Queensland Brisbane QLD 4072 Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland Brisbane QLD 4072 Australia
| | - Craig A Bell
- Centre for Advanced Imaging, The University of Queensland Brisbane QLD 4072 Australia .,Australian Institute for Bioengineering & Nanotechnology (AIBN), The University of Queensland Brisbane QLD 4072 Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland Brisbane QLD 4072 Australia
| | - Kristofer J Thurecht
- Centre for Advanced Imaging, The University of Queensland Brisbane QLD 4072 Australia .,Australian Institute for Bioengineering & Nanotechnology (AIBN), The University of Queensland Brisbane QLD 4072 Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland Brisbane QLD 4072 Australia
| |
Collapse
|
26
|
Rondon A, Degoul F. Antibody Pretargeting Based on Bioorthogonal Click Chemistry for Cancer Imaging and Targeted Radionuclide Therapy. Bioconjug Chem 2020; 31:159-173. [PMID: 31855602 DOI: 10.1021/acs.bioconjchem.9b00761] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bioorthogonal click chemistry-employing antibody-conjugated trans-cyclooctenes (TCO) and tetrazine (Tz)-based radioligands able to covalently bind in vivo-appeared recently as a potential alternative to circumvent the hematotoxicity induced by radioimmunotherapy of solid tumors. This Review focuses on the recent advances concerning TCO/Tz pretargeting in both cancer imaging and targeted-radionuclide therapy for prospective clinical transfer. We exhaustively identified 25 PubMed publications reporting preclinical imaging and 5 therapy studies with full mAbs as targeting vectors, since its first application in 2010. The fast, safe, modulable, and specific TCO/Tz pretargeting showed high potential as a theranostic tool to get more personalized and precise cancer care. The recent optimizations reported here highlighted a possible first clinical evaluation of IEDDA pretargeting in the coming years.
Collapse
Affiliation(s)
- Aurélie Rondon
- Université Clermont Auvergne , Imagerie Moléculaire et Stratégies Théranostiques , BP 184, F-63005 Clermont-Ferrand , France.,Inserm, U 1240 , F-63000 Clermont-Ferrand , France.,Centre Jean Perrin , F-63011 Clermont-Ferrand , France
| | - Françoise Degoul
- Université Clermont Auvergne , Imagerie Moléculaire et Stratégies Théranostiques , BP 184, F-63005 Clermont-Ferrand , France.,Inserm, U 1240 , F-63000 Clermont-Ferrand , France.,Centre Jean Perrin , F-63011 Clermont-Ferrand , France
| |
Collapse
|
27
|
Johann K, Svatunek D, Seidl C, Rizzelli S, Bauer TA, Braun L, Koynov K, Mikula H, Barz M. Tetrazine- and trans-cyclooctene-functionalised polypept(o)ides for fast bioorthogonal tetrazine ligation. Polym Chem 2020. [DOI: 10.1039/d0py00375a] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Tetrazine- and trans-cyclooctene-functionalised polypeptides and polypetoids were prepared by ring-opening polymerisation of N-carboxyanhydrides using the respective functional initiators and shown to react in fast bioorthogonal tetrazine ligations.
Collapse
Affiliation(s)
- Kerstin Johann
- Department of Chemistry
- Johannes Gutenberg University Mainz
- 55128 Mainz
- Germany
| | - Dennis Svatunek
- Institute of Applied Synthetic Chemistry
- Technische Universität Wien
- 1060 Vienna
- Austria
| | - Christine Seidl
- Department of Chemistry
- Johannes Gutenberg University Mainz
- 55128 Mainz
- Germany
| | - Silvia Rizzelli
- Department of Chemistry
- Johannes Gutenberg University Mainz
- 55128 Mainz
- Germany
| | - Tobias A. Bauer
- Department of Chemistry
- Johannes Gutenberg University Mainz
- 55128 Mainz
- Germany
| | - Lydia Braun
- Department of Chemistry
- Johannes Gutenberg University Mainz
- 55128 Mainz
- Germany
| | - Kaloian Koynov
- Max Planck Institute for Polymer Research
- 55128 Mainz
- Germany
| | - Hannes Mikula
- Institute of Applied Synthetic Chemistry
- Technische Universität Wien
- 1060 Vienna
- Austria
| | - Matthias Barz
- Department of Chemistry
- Johannes Gutenberg University Mainz
- 55128 Mainz
- Germany
| |
Collapse
|
28
|
Rondon A, Schmitt S, Briat A, Ty N, Maigne L, Quintana M, Membreno R, Zeglis BM, Navarro-Teulon I, Pouget JP, Chezal JM, Miot-Noirault E, Moreau E, Degoul F. Pretargeted radioimmunotherapy and SPECT imaging of peritoneal carcinomatosis using bioorthogonal click chemistry: probe selection and first proof-of-concept. Theranostics 2019; 9:6706-6718. [PMID: 31588245 PMCID: PMC6771248 DOI: 10.7150/thno.35461] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 06/25/2019] [Indexed: 12/26/2022] Open
Abstract
Rationale: Pretargeted radioimmunotherapy (PRIT) based upon bioorthogonal click chemistry has been investigated for the first time in the context of peritoneal carcinomatosis using a CEA-targeting 35A7 mAb bearing trans-cyclooctene (TCO) moieties and several 177Lu-labeled tetrazine (Tz) radioligands. Starting from three Tz probes containing PEG linkers of varying lengths between the DOTA and Tz groups (i.e. PEGn = 3, 7, or 11, respectively, for Tz-1, Tz-2, and Tz-3), we selected [177Lu]Lu-Tz-2 as the most appropriate for pretargeted SPECT imaging and demonstrated its efficacy in tumor growth control. Methods: An orthotopic model of peritoneal carcinomatosis (PC) was obtained following the intraperitoneal (i.p.) injection of A431-CEA-Luc cells in nude mice. Tumor growth was assessed using bioluminescence imaging. Anti-CEA 35A7 mAb was grafted with 2-3 TCO per immunoglobulin. Pretargeted SPECT imaging and biodistribution experiments were performed to quantify the activity concentrations of [177Lu]Lu-Tz-1-3 in tumors and non-target organs to determine the optimal Tz probe for the PRIT of PC. Results: The pharmacokinetic profiles of [177Lu]Lu-Tz-1-3 alone were determined using both SPECT imaging and biodistribution experiments. These data revealed that [177Lu]Lu-Tz-1 was cleared via both the renal and hepatic systems, while [177Lu]Lu-Tz-2 and [177Lu]Lu-Tz-3 were predominantly excreted via the renal system. In addition, these results illuminated that the longer the PEG linker, the more rapidly the Tz radioligand was cleared from the peritoneal cavity. The absorbed radiation dose corresponding to pretargeting with 35A7-TCO followed 24 h later by [177Lu]Lu-Tz-1-4 was higher for tumors following the administration of [177Lu]Lu-Tz-2 (i.e. 0.59 Gy/MBq) compared to either [177Lu]Lu-Tz-1 (i.e. 0.25 Gy/MBq) and [177Lu]Lu-Tz-3 (i.e. 0.18 Gy/MBq). In a longitudinal PRIT study, we showed that the i.p. injection of 40 MBq of [177Lu]Lu-Tz-2 24 hours after the systemic administration of 35A7-TCO significantly slowed tumor growth compared to control mice receiving only saline or 40 MBq of [177Lu]Lu-Tz-2 alone. Ex vivo measurement of the peritoneal carcinomatosis index (PCI) confirmed that PRIT significantly reduced tumor growth (PCI = 15.5 ± 2.3 after PRIT vs 30.0 ± 2.3 and 30.8 ± 1.4 for the NaCl and [177Lu]Lu-Tz-2 alone groups, respectively). Conclusion: Our results clearly demonstrate the impact of the length of PEG linkers upon the biodistribution profiles of 177Lu-labeled Tz radioligands. Furthermore, we demonstrated for the first time the possibility of using bioorthogonal chemistry for both the pretargeted SPECT and PRIT of peritoneal carcinomatosis.
Collapse
|
29
|
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
|
30
|
Therapeutic Applications of Pretargeting. Pharmaceutics 2019; 11:pharmaceutics11090434. [PMID: 31480515 PMCID: PMC6781323 DOI: 10.3390/pharmaceutics11090434] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/09/2019] [Accepted: 08/10/2019] [Indexed: 02/06/2023] Open
Abstract
Targeted therapies, such as radioimmunotherapy (RIT), present a promising treatment option for the eradication of tumor lesions. RIT has shown promising results especially for hematologic malignancies, but the therapeutic efficacy is limited by unfavorable tumor-to-background ratios resulting in high radiotoxicity. Pretargeting strategies can play an important role in addressing the high toxicity profile of RIT. Key to pretargeting is the concept of decoupling the targeting vehicle from the cytotoxic agent and administrating them separately. Studies have shown that this approach has the ability to enhance the therapeutic index as it can reduce side effects caused by off-target irradiation and thereby increase curative effects due to higher tolerated doses. Pretargeted RIT (PRIT) has been explored for imaging and treatment of different cancer types over the years. This review will give an overview of the various targeted therapies in which pretargeting has been applied, discussing PRIT with alpha- and beta-emitters and as part of combination therapy, plus its use in drug delivery systems.
Collapse
|
31
|
Pretargeted Nuclear Imaging and Radioimmunotherapy Based on the Inverse Electron-Demand Diels-Alder Reaction and Key Factors in the Pretargeted Synthetic Design. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2019:9182476. [PMID: 31531006 PMCID: PMC6732628 DOI: 10.1155/2019/9182476] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/20/2019] [Accepted: 08/01/2019] [Indexed: 11/18/2022]
Abstract
The exceptional speed and biorthogonality of the inverse electron-demand Diels-Alder (IEDDA) click chemistry between 1,2,4,5-tetrazines and strained alkene dienophiles have made it promising in the realm of pretargeted imaging and therapy. During the past 10 years, the IEDDA-pretargeted strategies have been tested and have already proven capable of producing images with high tumor-to-background ratios and improving therapeutic effect. This review will focus on recent applications of click chemistry ligations in the pretargeted imaging studies of single photon emission computed tomography (SPECT), positron emission tomography (PET), and pretargeted radioimmunotherapy investigations. Additionally, the influence factors of stability, reactivity, and pharmacokinetic properties of TCO tag modified immunoconjugates and radiolabeled Tz derivatives were also summarized in this article, which should be carefully considered in the system design in order to develop a successful pretargeted methodology. We hope that this review will not only equip readers with a knowledge of pretargeted methodology based on IEDDA click chemistry but also inspire synthetic chemists and radiochemists to develop pretargeted radiopharmaceutical components in a more innovative way with various influence factors considered.
Collapse
|
32
|
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
|
33
|
Membreno R, Cook BE, Zeglis BM. Pretargeted Radioimmunotherapy Based on the Inverse Electron Demand Diels-Alder Reaction. J Vis Exp 2019. [PMID: 30774125 DOI: 10.3791/59041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
While radioimmunotherapy (RIT) is a promising approach for the treatment of cancer, the long pharmacokinetic half-life of radiolabeled antibodies can result in high radiation doses to healthy tissues. Perhaps not surprisingly, several different strategies have been developed to circumvent this troubling limitation. One of the most promising of these approaches is pretargeted radioimmunotherapy (PRIT). PRIT is predicated on decoupling the radionuclide from the immunoglobulin, injecting them separately, and then allowing them to combine in vivo at the target tissue. This approach harnesses the exceptional tumor-targeting properties of antibodies while skirting their pharmacokinetic drawbacks, thereby lowering radiation doses to non-target tissues and facilitating the use of radionuclides with half-lives that are considered too short for use in traditional radioimmunoconjugates. Over the last five years, our laboratory and others have developed an approach to in vivo pretargeting based on the inverse electron-demand Diels-Alder (IEDDA) reaction between trans-cyclooctene (TCO) and tetrazine (Tz). This strategy has been successfully applied to pretargeted positron emission tomography (PET) and single-photon emission computed tomography (SPECT) imaging with a variety of antibody-antigen systems. In a pair of recent publications, we have demonstrated the efficacy of IEDDA-based PRIT in murine models of pancreatic ductal adenocarcinoma and colorectal carcinoma. In this protocol, we describe protocols for PRIT using a 177Lu-DOTA-labeled tetrazine radioligand ([177Lu]Lu-DOTA-PEG7-Tz) and a TCO-modified variant of the colorectal cancer targeting huA33 antibody (huA33-TCO). More specifically, we will describe the construction of huA33-TCO, the synthesis and radiolabeling of [177Lu]Lu-DOTA-PEG7-Tz, and the performance of in vivo biodistribution and longitudinal therapy studies in murine models of colorectal carcinoma.
Collapse
Affiliation(s)
- Rosemery Membreno
- Department of Chemistry, Hunter College of the City University of New York; Ph.D. Program in Chemistry, Graduate Center of the City University of New York
| | - Brendon E Cook
- Department of Chemistry, Hunter College of the City University of New York; Ph.D. Program in Chemistry, Graduate Center of the City University of New York; Department of Radiology, Memorial Sloan Kettering Cancer Center
| | - Brian M Zeglis
- Department of Chemistry, Hunter College of the City University of New York; Ph.D. Program in Chemistry, Graduate Center of the City University of New York; Department of Radiology, Memorial Sloan Kettering Cancer Center; Department of Radiology, Weill Cornell Medical College;
| |
Collapse
|
34
|
Poty S, Carter LM, Mandleywala K, Membreno R, Abdel-Atti D, Ragupathi A, Scholz WW, Zeglis BM, Lewis JS. Leveraging Bioorthogonal Click Chemistry to Improve 225Ac-Radioimmunotherapy of Pancreatic Ductal Adenocarcinoma. Clin Cancer Res 2019; 25:868-880. [PMID: 30352909 PMCID: PMC6343144 DOI: 10.1158/1078-0432.ccr-18-1650] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 08/28/2018] [Accepted: 10/18/2018] [Indexed: 12/25/2022]
Abstract
PURPOSE Interest in targeted alpha-therapy has surged due to α-particles' high cytotoxicity. However, the widespread clinical use of this approach could be limited by on-/off-target toxicities. Here, we investigated the inverse electron-demand Diels-Alder ligation between an 225Ac-labeled tetrazine radioligand and a trans-cyclooctene-bearing anti-CA19.9 antibody (5B1) for pretargeted α-radioimmunotherapy (PRIT) of pancreatic ductal adenocarcinoma (PDAC). This alternative strategy is expected to reduce nonspecific toxicities as compared with conventional radioimmunotherapy (RIT).Experimental Design: A side-by-side comparison of 225Ac-PRIT and conventional RIT using a directly 225Ac-radiolabeled immunoconjugate evaluates the therapeutic efficacy and toxicity of both methodologies in PDAC murine models. RESULTS A comparative biodistribution study of the PRIT versus RIT methodology underscored the improved pharmacokinetic properties (e.g., prolonged tumor uptake and increased tumor-to-tissue ratios) of the PRIT approach. Cerenkov imaging coupled to PRIT confirmed the in vivo biodistribution of 225Ac-radioimmunoconjugate but-importantly-further allowed for the ex vivo monitoring of 225Ac's radioactive daughters' redistribution. Human dosimetry was extrapolated from the mouse biodistribution and confirms the clinical translatability of 225Ac-PRIT. Furthermore, longitudinal therapy studies performed in subcutaneous and orthotopic PDAC models confirm the therapeutic efficacy of 225Ac-PRIT with the observation of prolonged median survival compared with control cohorts. Finally, a comparison with conventional RIT highlighted the potential of 225Ac-PRIT to reduce hematotoxicity while maintaining therapeutic effectiveness. CONCLUSIONS The ability of 225Ac-PRIT to deliver a radiotherapeutic payload while simultaneously reducing the off-target toxicity normally associated with RIT suggests that the clinical translation of this approach will have a profound impact on PDAC therapy.
Collapse
Affiliation(s)
- Sophie Poty
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Lukas M Carter
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Komal Mandleywala
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rosemery Membreno
- Department of Chemistry, Hunter College of the City University of New York, New York, New York
- Ph.D. Program in Chemistry, Graduate Center of the City University of New York, New York, New York
| | - Dalya Abdel-Atti
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ashwin Ragupathi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Brian M Zeglis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.
- Department of Chemistry, Hunter College of the City University of New York, New York, New York
- Ph.D. Program in Chemistry, Graduate Center of the City University of New York, New York, New York
- Departments of Radiology and Department of Pharmacology, Weill Cornell Medical College, New York, New York
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.
- Departments of Radiology and Department of Pharmacology, Weill Cornell Medical College, New York, New York
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
35
|
Liu G. A Revisit to the Pretargeting Concept-A Target Conversion. Front Pharmacol 2018; 9:1476. [PMID: 30618765 PMCID: PMC6304396 DOI: 10.3389/fphar.2018.01476] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 12/03/2018] [Indexed: 01/22/2023] Open
Abstract
Pretargeting is often used as a tumor targeting strategy that provides much higher tumor to non-tumor ratios than direct-targeting using radiolabeled antibody. Due to the multiple injections, pretargeting is investigated less than direct targeting, but the high T/NT ratios have rendered it more useful for therapy. While the progress in using this strategy for tumor therapy has been regularly reviewed in the literature, this review focuses on the nature and quantitative understanding of the pretargeting concept. By doing so, it is the goal of this review to accelerate pretargeting development and translation to the clinic and to prepare the researchers who are not familiar with the pretargeting concept but are interested in applying it. The quantitative understanding is presented in a way understandable to the average researchers in the areas of drug development and clinical translation who have the basic concept of calculus and general chemistry.
Collapse
Affiliation(s)
- Guozheng Liu
- Department of Radiology, University of Massachusetts Medical School Worcester, MA, United States
| |
Collapse
|
36
|
Abstract
Radiometals possess an exceptional breadth of decay properties and have been applied to medicine with great success for several decades. The majority of current clinical use involves diagnostic procedures, which use either positron-emission tomography (PET) or single-photon imaging to detect anatomic abnormalities that are difficult to visualize using conventional imaging techniques (e.g., MRI and X-ray). The potential of therapeutic radiometals has more recently been realized and relies on ionizing radiation to induce irreversible DNA damage, resulting in cell death. In both cases, radiopharmaceutical development has been largely geared toward the field of oncology; thus, selective tumor targeting is often essential for efficacious drug use. To this end, the rational design of four-component radiopharmaceuticals has become popularized. This Review introduces fundamental concepts of drug design and applications, with particular emphasis on bifunctional chelators (BFCs), which ensure secure consolidation of the radiometal and targeting vector and are integral for optimal drug performance. Also presented are detailed accounts of production, chelation chemistry, and biological use of selected main group and rare earth radiometals.
Collapse
Affiliation(s)
- Thomas I Kostelnik
- Medicinal Inorganic Chemistry Group, Department of Chemistry , University of British Columbia , Vancouver , British Columbia V6T 1Z1 , Canada
| | - Chris Orvig
- Medicinal Inorganic Chemistry Group, Department of Chemistry , University of British Columbia , Vancouver , British Columbia V6T 1Z1 , Canada
| |
Collapse
|
37
|
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
|