1
|
Kang CM, Kim JL, Jung HJ, Jung KH, Kim M, Kim G, Lee H, Lee KH. 177Lu-Anti-CD25 Antibody for Interleukin-2 Receptor-α-Targeted Radioimmunotherapy of SUDHL1 Lymphomas in Mice. Mol Pharm 2025. [PMID: 40408541 DOI: 10.1021/acs.molpharmaceut.4c01410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Antibodies (Abs) conjugated with particle-emitting radioisotopes are used for cancer therapy, and the CD25 receptor (IL-2Rα) is a promising target for lymphomas. We functionalized an anti-CD25 Ab with TCO-PEG3-maleimide, specifically on sulfhydryl moieties, at 1.89 molecules per Ab. Radiosynthesis was achieved by efficiently prelabeling DOTA-PEG4-tetrazine with 177Lu at a high temperature and then linking it to the TCO-conjugated Abs via facile click chemistry under mild conditions. The [177Lu]Lu-DOTA-PEG4-Tz-TCO-PEG3-anti-CD25 Ab (177Lu-CD25 Ab) had a radiochemical purity of >99%, a specific activity of 707.9 ± 271.5 MBq/mg, an immunoreactive fraction of 77.6%, and high radiolabel stability in serum for up to 7 days. CD25-positive SUDHL1 human T lymphoma cells showed 177Lu-CD25 Ab uptake that was completely blocked by pretreatment with unlabeled Ab. The 177Lu-CD25 Ab dose-dependently suppressed SUDHL1 cell survival in vitro. In mice, 177Lu-CD25 Ab uptake at 5 days was high in the SUDHL1 tumors (7.1 ± 1.6%ID/g), modest in the liver, kidneys, and spleen, and low in the blood, lungs, and bones. CD25-specific targeting was confirmed by 66.7% suppression of tumor uptake by pretreatment with unlabeled CD25 Ab. Treatment with 18.5 MBq of 177Lu-CD25 Ab shrank the xenograft tumors, and they remained undetectable until study termination on day 61. In contrast, the tumors in all control and CD25 Ab-treated mice grew to exceed the end point criterion of 2,000 mm3. The standardized tumor growth rate and 19-day tumor volume were completely suppressed in the 177Lu-CD25 Ab group (109.9 ± 73.5 and 132.6 ± 111.5 mm3), compared with the control (1053.9 ± 151.1 and 1804.5 ± 283.1 mm3) and CD25 Ab groups (1049.7 ± 212.2 and 1443.8 ± 839.4 mm3; all p < 0.001). A Kaplan-Meier survival analysis showed that 177Lu-CD25 Ab-treated mice survived significantly longer than mice in the control and CD25 Ab groups. Tumors in a separate set of mice that were treated with 177Lu-CD25 Ab displayed increased PARP1 cleavage fragments, a signature of apoptosis. Toxicity studies showed that white blood cell, red blood cell, and platelet counts in the 177Lu-CD25 Ab group decreased from days 3-14, reaching a nadir on days 17-21 and returning to the normal range by days 24-31. Liver and renal function tests on day 28 did not differ from those of untreated mice. Thus, the 177Lu-CD25 Ab prepared as described here could be useful for radioimmunotherapy of CD25-positive lymphomas.
Collapse
Affiliation(s)
- Choong Mo Kang
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea
- Radiological and Medico-Oncological Sciences, University of Science and Technology, Seoul 01812, Korea
| | - Jung Lim Kim
- Department of Nuclear Medicine, Samsung Medical Center, Seoul 06351, Korea
- Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University School of Medicine, Seoul 06355, Korea
| | - Hye Jin Jung
- Department of Nuclear Medicine, Samsung Medical Center, Seoul 06351, Korea
| | - Kyung-Ho Jung
- Department of Nuclear Medicine, Samsung Medical Center, Seoul 06351, Korea
- Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University School of Medicine, Seoul 06355, Korea
| | - Mina Kim
- Department of Nuclear Medicine, Samsung Medical Center, Seoul 06351, Korea
- Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University School of Medicine, Seoul 06355, Korea
| | - Giro Kim
- Department of Nuclear Medicine, Samsung Medical Center, Seoul 06351, Korea
| | - Hyunjong Lee
- Department of Nuclear Medicine, Samsung Medical Center, Seoul 06351, Korea
| | - Kyung-Han Lee
- Department of Nuclear Medicine, Samsung Medical Center, Seoul 06351, Korea
- Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University School of Medicine, Seoul 06355, Korea
| |
Collapse
|
2
|
Wehrmüller JE, Frei JC, Hechler T, Kulke M, Pahl A, Béhé M, Schibli R, Spycher PR. Site-Specific Modification of Native IgGs with Flexible Drug-Load. Chembiochem 2025; 26:e202400511. [PMID: 39305147 PMCID: PMC12007069 DOI: 10.1002/cbic.202400511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/11/2024] [Indexed: 11/08/2024]
Abstract
Homogeneous, site-specifically conjugated antibodies have shown to result in antibody-drug conjugates (ADCs) with improved efficacy and tolerability compared to stochastically conjugated ADCs. However, precisely controlling the drug load as well as attaching multiple payload moieties to the antibody remains challenging. Here, we demonstrate the simple and direct modification of native IgG-antibodies at the residue glutamine 295 (Q295) without the need for any protein engineering with flexible drug-to-antibody ratios of one or multiple payloads. The conjugation is enabled through short, positively charged lysine containing peptides and native, commercially available microbial transglutaminase. In proof-of-concept studies, HER2-targeting ADCs based on trastuzumab were generated with drug-to-antibody ratios (DARs) of 2 and 4 of the same or different payloads using orthogonal conjugation chemistries. Quantitative biodistribution studies performed with 111In-radiolabeled conjugates showed high tumour uptake and low accumulation of radioactivity in non-targeted tissues. A single dose study of trastuzumab conjugated to the highly potent payload α-Amanitin demonstrated complete and long-lasting tumour remission and was well-tolerated at all dose levels tested.
Collapse
Affiliation(s)
- Jöri E. Wehrmüller
- Center for Radiopharmaceutical ScienceETH-PSI-USZPaul Scherrer Institute5232Villigen-PSISwitzerland
| | - Julia C. Frei
- Center for Radiopharmaceutical ScienceETH-PSI-USZPaul Scherrer Institute5232Villigen-PSISwitzerland
| | - Torsten Hechler
- Heidelberg Pharma Research GmbHGregor-Mendel-Straße 2268526LadenburgGermany
| | - Michael Kulke
- Heidelberg Pharma Research GmbHGregor-Mendel-Straße 2268526LadenburgGermany
| | - Andreas Pahl
- Heidelberg Pharma Research GmbHGregor-Mendel-Straße 2268526LadenburgGermany
| | - Martin Béhé
- Center for Radiopharmaceutical ScienceETH-PSI-USZPaul Scherrer Institute5232Villigen-PSISwitzerland
| | - Roger Schibli
- Center for Radiopharmaceutical ScienceETH-PSI-USZPaul Scherrer Institute5232Villigen-PSISwitzerland
- Department of Chemistry and Applied BiosciencesETH Zürich8093ZürichSwitzerland
| | - Philipp R. Spycher
- Center for Radiopharmaceutical ScienceETH-PSI-USZPaul Scherrer Institute5232Villigen-PSISwitzerland
- Araris Biotech AGRiedhofstrasse 118804Au ZHSwitzerland
| |
Collapse
|
3
|
Fayn S, Roy S, Cabalteja CC, Lee W, Makala H, Baidoo K, Nambiar D, Sheehan‐Klenk J, Chung J, Buffington J, Ho M, Escorcia FE, Cheloha RW. Generation of Site-Specifically Labeled Affinity Reagents via Use of a Self-Labeling Single Domain Antibody. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417160. [PMID: 39965119 PMCID: PMC11984916 DOI: 10.1002/advs.202417160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/30/2025] [Indexed: 02/20/2025]
Abstract
Several chemical and enzymatic methods have been used to link antibodies to moieties that facilitate visualization of cognate targets. Emerging evidence suggests that the extent of labeling, dictated by the type of chemistry used, has a substantial impact on performance, especially in the context of antibodies used for the visualization of tumors in vivo. These effects are particularly pronounced in studies using small antibody fragments, such as single-domain antibodies, or nanobodies. Here, we leverage a new variety of conjugation chemistry, based on a nanobody that forms a crosslink with a specialized high-affinity epitope analogue, to label target-specific nanobody constructs with functionalities of choice, including fluorophores, chelators, and click chemistry handles. Using heterodimeric nanobody conjugates, comprised of an antigen recognition module and a self-labeling module, enables us to attach the desired functional group at a location distal to the site of antigen recognition. Constructs generated using this approach bound to antigens expressed on xenograft murine models of liver cancer and allowed for non-invasive diagnostic imaging. The modularity of our approach using a self-labeling nanobody offers a novel method for site-specific functionalization of biomolecules and can be extended to other applications for which covalent labeling is required.
Collapse
Affiliation(s)
- Stanley Fayn
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
- Oxford Institute for Radiation OncologyDepartment of OncologyUniversity of OxfordOxfordOX3 7DQUK
| | - Swarnali Roy
- Chemical Biology in Signaling SectionNational Institutes of Diabetes and Digestive and Kidney DiseasesNational Institutes of HealthBethesdaMD20892USA
| | - Chino C. Cabalteja
- Chemical Biology in Signaling SectionNational Institutes of Diabetes and Digestive and Kidney DiseasesNational Institutes of HealthBethesdaMD20892USA
| | - Woonghee Lee
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Hima Makala
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Kwamena Baidoo
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Divya Nambiar
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Julia Sheehan‐Klenk
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Joon‐Yong Chung
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Jesse Buffington
- Antibody Engineering ProgramCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Mitchell Ho
- Antibody Engineering ProgramCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
- Laboratory of Molecular BiologyCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Freddy E. Escorcia
- Molecular Imaging BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
- Radiation Oncology BranchCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20892USA
| | - Ross W. Cheloha
- Chemical Biology in Signaling SectionNational Institutes of Diabetes and Digestive and Kidney DiseasesNational Institutes of HealthBethesdaMD20892USA
| |
Collapse
|
4
|
Thanasi IA, Bouloc N, McMahon C, Wang N, Szijj PA, Butcher T, Rochet LNC, Love EA, Merritt A, Baker JR, Chudasama V. Formation of mono- and dual-labelled antibody fragment conjugates via reversible site-selective disulfide modification and proximity induced lysine reactivity. Chem Sci 2025; 16:2763-2776. [PMID: 39811008 PMCID: PMC11726237 DOI: 10.1039/d4sc06500j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Many protein bioconjugation strategies focus on the modification of lysine residues owing to the nucleophilicity of their amine side-chain, the generally high abundance of lysine residues on a protein's surface and the ability to form robustly stable amide-based bioconjugates. However, the plethora of solvent accessible lysine residues, which often have similar reactivity, is a key inherent issue when searching for regioselectivity and/or controlled loading of an entity. A relevant example is the modification of antibodies and/or antibody fragments, whose conjugates offer potential for a wide variety of applications. Thus, research in this area for the controlled loading of an entity via reaction with lysine residues is of high importance. In this article, we present an approach to achieve this by exploiting the quantitative and reversible site-selective modification of disulfides using pyridazinediones, which facilitates near-quantitative proximity-induced reactions with lysines to enable controlled loading of an entity. The strategy was appraised on several clinically relevant antibody fragments and enabled the formation of mono-labelled lysine-modified antibody fragment conjugates via the formation of stable amide bonds and the use of click chemistry for modular modification. Furthermore, through the use of multiple cycles of this novel strategy, an orthogonally bis-labelled lysine-modified antibody fragment conjugate was also furnished.
Collapse
Affiliation(s)
- Ioanna A Thanasi
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Nathalie Bouloc
- LifeArc, Accelerator Building Open Innovation Campus Stevenage SG1 2FX UK
| | - Clíona McMahon
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Ning Wang
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Peter A Szijj
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Tobias Butcher
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Léa N C Rochet
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Elizabeth A Love
- LifeArc, Accelerator Building Open Innovation Campus Stevenage SG1 2FX UK
| | - Andy Merritt
- LifeArc, Accelerator Building Open Innovation Campus Stevenage SG1 2FX UK
| | - James R Baker
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Vijay Chudasama
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| |
Collapse
|
5
|
Ma C, Xu J, Wang X, Wang X, Zhang L, Jing S. Selenenylsulfide covalent-directed chemistry for the detection of sulfhydryl groups using a diselenide fluorescent probe. RSC Adv 2024; 14:36754-36762. [PMID: 39559574 PMCID: PMC11571120 DOI: 10.1039/d4ra05923a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024] Open
Abstract
We report the development of a diglycosyldiselenide-based fluorescent probe for the rapid detection of sulfhydryl-containing biomolecules. The probe facilitates a chemoselective coupling reaction with sulfhydryl groups in aqueous buffer under ambient conditions, resulting in the formation of homogeneous Se-S conjugates within one hour. Using glutathione, a sulfhydryl-containing biomolecule, as a proof of concept, the probe achieved a detection limit of 0.75 μM based on the 3σ criterion. The method was further extended to the fluorescent labeling of cysteine-containing peptides, proteins, and living bacterial cells, showcasing the utility of Se-S covalent-directed chemistry as an analytical tool. This approach underscores the considerable potential of diglycosyldiselenide-based fluorescent probes for broader applications in biochemical research.
Collapse
Affiliation(s)
- Chunqiu Ma
- School of Chemistry and Molecular Engineering, Nanjing Tech University 30 South Puzhu Road Nanjing 211816 Jiangsu China
| | - Jichao Xu
- School of Chemistry and Molecular Engineering, Nanjing Tech University 30 South Puzhu Road Nanjing 211816 Jiangsu China
| | - Xiaolu Wang
- School of Chemistry and Molecular Engineering, Nanjing Tech University 30 South Puzhu Road Nanjing 211816 Jiangsu China
| | - Xuewen Wang
- School of Chemistry and Molecular Engineering, Nanjing Tech University 30 South Puzhu Road Nanjing 211816 Jiangsu China
| | - Lei Zhang
- School of Chemistry and Molecular Engineering, Nanjing Tech University 30 South Puzhu Road Nanjing 211816 Jiangsu China
| | - Su Jing
- School of Chemistry and Molecular Engineering, Nanjing Tech University 30 South Puzhu Road Nanjing 211816 Jiangsu China
| |
Collapse
|
6
|
Rodriguez C, Sarrett SM, Sebastiano J, Delaney S, McGlone SA, Hosny MM, Thau S, Bournazos S, Zeglis BM. Exploring the Interplay Between Radioimmunoconjugates and Fcγ Receptors in Genetically Engineered Mouse Models of Cancer. ACS Pharmacol Transl Sci 2024; 7:3452-3461. [PMID: 39539260 PMCID: PMC11555515 DOI: 10.1021/acsptsci.4c00275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024]
Abstract
Fcγ receptors (FcγR) are responsible for many of the interactions between immunoglobulins (IgG) and immune cells. In biomedicine, this interplay is critical to the activity of several types of immunotherapeutics; however, relatively little is known about how FcγRs affect the in vivo performance of radiolabeled antibodies. A handful of recent preclinical studies suggest that binding by FcγR-and particularly FcγRI-can affect the pharmacokinetic profiles of 89Zr-labeled radioimmunoconjugates, but there are no extant studies in immunocompetent or genetically engineered mouse models of cancer. In the investigation at hand, we synthesized and characterized 89Zr-labeled probes based on wild-type and aglycosylated variants of the CA19-9-targeting antibody 5B1 and evaluated their in vivo behavior in several murine models of cancer, including immunocompetent and FcγR-humanized mice. The aglycosylated desferrioxamine (DFO)-bearing immunoconjugate DFO-N297A5B1 displayed identical binding to CA19-9-expressing cells compared to its wild-type analogue (DFO-5B1) but exhibited dramatically attenuated affinity for several FcγR. Positron emission tomography imaging and biodistribution studies with [89Zr]Zr-DFO-5B1 and [89Zr]Zr-DFO-N297A5B1 were subsequently performed in several strains of mice bearing CA19-9-expressing BxPC3 human pancreatic ductal adenocarcinoma and B16F10-FUT3 murine melanoma xenografts. Significant differences in the pharmacokinetics of the two radioimmunoconjugates were observed in tumor-bearing immunocompromised NSG mice, but these differences failed to materialize in immunocompetent C57BL/6 and FcγR-humanized C57BL/6 mice with B16F10-FUT3 xenografts. We hypothesize that these observations are related to the presence or absence of endogenous IgG. NSG mice completely lack endogenous IgG, and thus their mFcγR are free to bind radioimmunoconjugates and alter their pharmacokinetic behavior. In contrast, C57BL/6 and FcγR-humanized C57BL/6 mice both have endogenous IgG that occupy their FcγR (murine for the former and human for the latter), precluding interactions with radioimmunoconjugates. Ultimately, these data suggest that understanding the interplay between radiolabeled antibodies and FcγR is critical during the preclinical evaluation of radioimmunoconjugates.
Collapse
Affiliation(s)
- Cindy Rodriguez
- Department
of Chemistry, Hunter College, City University
of New York, New York 10021, New York, United States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York 10021, New York, United States
- Ph.D.
Program in Chemistry, Graduate Center of
City University of New York, New
York 10021, New York, United States
| | - Samantha M. Sarrett
- Department
of Chemistry, Hunter College, City University
of New York, New York 10021, New York, United States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York 10021, New York, United States
- Ph.D.
Program in Biochemistry, Graduate Center
of City University of New York, New York 10021, New
York, United States
| | - Joni Sebastiano
- Department
of Chemistry, Hunter College, City University
of New York, New York 10021, New York, United States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York 10021, New York, United States
- Ph.D.
Program in Biochemistry, Graduate Center
of City University of New York, New York 10021, New
York, United States
| | - Samantha Delaney
- Department
of Chemistry, Hunter College, City University
of New York, New York 10021, New York, United States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York 10021, New York, United States
- Ph.D.
Program in Biochemistry, Graduate Center
of City University of New York, New York 10021, New
York, United States
| | - Shane A. McGlone
- Department
of Chemistry, Hunter College, City University
of New York, New York 10021, New York, United States
| | - Meena M. Hosny
- Department
of Chemistry, Hunter College, City University
of New York, New York 10021, New York, United States
| | - Sarah Thau
- Department
of Chemistry, Hunter College, City University
of New York, New York 10021, New York, United States
| | - Stylianos Bournazos
- Laboratory
of Molecular Genetics and Immunology, The
Rockefeller University, 1230 York Avenue, New York 10065, New York, United States
| | - Brian M. Zeglis
- Department
of Chemistry, Hunter College, City University
of New York, New York 10021, New York, United States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York 10021, New York, United States
- Ph.D.
Program in Chemistry, Graduate Center of
City University of New York, New
York 10021, New York, United States
- Ph.D.
Program in Biochemistry, Graduate Center
of City University of New York, New York 10021, New
York, United States
- Department
of Radiology, Weill Cornell Medical College, New York 10021, New York, United States
| |
Collapse
|
7
|
Vanermen M, Ligeour M, Oliveira MC, Gestin JF, Elvas F, Navarro L, Guérard F. Astatine-211 radiolabelling chemistry: from basics to advanced biological applications. EJNMMI Radiopharm Chem 2024; 9:69. [PMID: 39365487 PMCID: PMC11452365 DOI: 10.1186/s41181-024-00298-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/17/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND 211At-radiopharmaceuticals are currently the subject of growing studies for targeted alpha therapy of cancers, which leads to the widening of the scope of the targeting vectors, from small molecules to peptides and proteins. This has prompted, during the past decade, to a renewed interest in developing novel 211At-labelling approaches and novel prosthetic groups to address the diverse scenarios and to reach improved efficiency and robustness of procedures as well as an appropriate in vivo stability of the label. MAIN BODY Translated from the well-known (radio)iodine chemistry, the long preferred electrophilic astatodemetallation using trialkylaryltin precursors is now complemented by new approaches using electrophilic or nucleophilic At. Alternatives to the astatoaryl moiety have been proposed to improve labelling stability, and the range of prosthetic groups available to label proteins has expanded. CONCLUSION In this report, we cover the evolution of radiolabelling chemistry, from the initial strategies developed in the late 1970's to the most recent findings.
Collapse
Affiliation(s)
- Maarten Vanermen
- Molecular Imaging and Radiology (MIRA), University of Antwerp, Wilrijk, Belgium
| | - Mathilde Ligeour
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - Maria-Cristina Oliveira
- Departamento de Engenharia e Ciências Nucleares and Centro de Ciências e Tecnologias Nucleares, IST, Universidade de Lisboa, Estrada Nacional 10 (km 139,7), 2695-066, Bobadela LRS, Portugal
| | | | - Filipe Elvas
- Molecular Imaging and Radiology (MIRA), University of Antwerp, Wilrijk, Belgium
| | | | - François Guérard
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France.
| |
Collapse
|
8
|
Sebastiano J, Samuels ZV, Kao WS, Zeglis BM. Site-specific bioconjugation and nuclear imaging. Curr Opin Chem Biol 2024; 81:102471. [PMID: 38833913 PMCID: PMC11323144 DOI: 10.1016/j.cbpa.2024.102471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/15/2024] [Accepted: 05/08/2024] [Indexed: 06/06/2024]
Abstract
Monoclonal antibodies and antibody fragments have proven to be highly effective vectors for the delivery of radionuclides to target tissues for positron emission tomography (PET) and single-photon emission computed tomography (SPECT). However, the stochastic methods that have traditionally been used to attach radioisotopes to these biomolecules inevitably produce poorly defined and heterogeneous probes and can impair the ability of the immunoglobulins to bind their molecular targets. In response to this challenge, an array of innovative site-specific and site-selective bioconjugation strategies have been developed, and these approaches have repeatedly been shown to yield better-defined and more homogeneous radioimmunoconjugates with superior in vivo performance than their randomly modified progenitors. In this Current Opinion in Chemical Biology review, we will examine recent advances in this field, including the development - and, in some cases, clinical translation - of nuclear imaging agents radiolabeled using strategies that target the heavy chain glycans, peptide tags, and unnatural amino acids.
Collapse
Affiliation(s)
- Joni Sebastiano
- 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 Biochemistry, Graduate Center of City University of New York, New York, NY, USA
| | - Zachary V Samuels
- 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 City University of New York, New York, NY, USA
| | - Wei-Siang Kao
- 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
| | - Brian M Zeglis
- 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 Biochemistry, Graduate Center of City University of New York, New York, NY, USA; Ph.D. Program in Chemistry, Graduate Center of City University of New York, New York, NY, USA; Department of Radiology, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
9
|
Badier L, Quelven I. Zirconium 89 and Copper 64 for ImmunoPET: From Antibody Bioconjugation and Radiolabeling to Molecular Imaging. Pharmaceutics 2024; 16:882. [PMID: 39065579 PMCID: PMC11279968 DOI: 10.3390/pharmaceutics16070882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/15/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Immunotherapy has transformed cancer treatment. Nevertheless, given the heterogeneity of clinical efficacy, the multiplicity of treatment options available and the possibility of serious adverse effects, selecting the most effective treatment has become the greatest challenge. Molecular imaging offers an attractive way for this purpose. ImmunoPET provides specific imaging with positron emission tomography (PET) using monoclonal antibodies (mAb) or its fragments as vector. By combining the high targeting specificity of mAb and the sensitivity of PET technique, immunoPET could noninvasively and dynamically reveal tumor antigens expression and provide theranostic tools of several types of malignancies. Because of their slow kinetics, mAbs require radioelements defined by a consistent half-life. Zirconium 89 (89Zr) and Copper 64 (64Cu) are radiometals with half-lives suitable for mAb labeling. Radiolabeling with a radiometal requires the prior use of a bifunctional chelate agent (BFCA) to functionalize mAb for radiometal chelation, in a second step. There are a number of BFCA available and much research is focused on antibody functionalization techniques or on developing the optimum chelating agent depending the selected radiometal. In this manuscript, we present a critical account of radiochemical techniques with radionuclides 89Zr and 64Cu and their applications in preclinical and clinical immuno-PET imaging.
Collapse
Affiliation(s)
| | - Isabelle Quelven
- Toulouse NeuroImaging Center (ToNIC), INSERM/UPS UMR 1214, University Hospital of Toulouse-Purpan, CEDEX 3, 31024 Toulouse, France;
| |
Collapse
|
10
|
Guo QR, Cao YJ. Applications of genetic code expansion technology in eukaryotes. Protein Cell 2024; 15:331-363. [PMID: 37847216 PMCID: PMC11074999 DOI: 10.1093/procel/pwad051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/26/2023] [Indexed: 10/18/2023] Open
Abstract
Unnatural amino acids (UAAs) have gained significant attention in protein engineering and drug development owing to their ability to introduce new chemical functionalities to proteins. In eukaryotes, genetic code expansion (GCE) enables the incorporation of UAAs and facilitates posttranscriptional modification (PTM), which is not feasible in prokaryotic systems. GCE is also a powerful tool for cell or animal imaging, the monitoring of protein interactions in target cells, drug development, and switch regulation. Therefore, there is keen interest in utilizing GCE in eukaryotic systems. This review provides an overview of the application of GCE in eukaryotic systems and discusses current challenges that need to be addressed.
Collapse
Affiliation(s)
- Qiao-ru Guo
- State Key Laboratory of Chemical Oncogenomic, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Yu J Cao
- State Key Laboratory of Chemical Oncogenomic, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China
| |
Collapse
|
11
|
Delaney S, Grimaldi C, Houghton JL, Zeglis BM. MIB Guides: Measuring the Immunoreactivity of Radioimmunoconjugates. Mol Imaging Biol 2024; 26:213-221. [PMID: 38446323 PMCID: PMC10973015 DOI: 10.1007/s11307-024-01898-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/04/2024] [Accepted: 01/24/2024] [Indexed: 03/07/2024]
Abstract
Immunoglobulins, both full-length antibodies and smaller antibody fragments, have long been regarded as effective platforms for diagnostic and therapeutic radiopharmaceuticals. The construction of radiolabeled immunoglobulins (i.e., radioimmunoconjugates) requires the manipulation of the biomolecule through the attachment of a radiohalogen or the bioconjugation of a chelator that is subsequently used to coordinate a radiometal. Both synthetic approaches have historically relied upon the stochastic modification of amino acids within the immunoglobulin, a process which poses a risk to the structural and functional integrity of the biomolecule itself. Not surprisingly, radioimmunoconjugates with impaired antigen binding capacity will inevitably exhibit suboptimal in vivo performance. As a result, the biological characterization of any newly synthesized radioimmunoconjugate must include an assessment of whether it has retained its ability to bind its antigen. Herein, we provide straightforward and concise protocols for three assays that can be used to determine the immunoreactivity of a radioimmunoconjugate: (1) a cell-based linear extrapolation assay; (2) a cell-based antigen saturation assay; and (3) a resin- or bead-based assay. In addition, we will provide a critical analysis of the relative merits of each assay, an examination of the inherent limitations of immunoreactivity assays in general, and a discussion of other approaches that may be used to interrogate the biological behavior of radioimmunoconjugates.
Collapse
Affiliation(s)
- Samantha Delaney
- Department of Chemistry, Hunter College of the City University of New York, New York, NY, USA
- Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Camilla Grimaldi
- Department of Chemistry, Hunter College of the City University of New York, New York, NY, USA
- Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jacob L Houghton
- Department of Radiology, State University of New York at Stony Brook, 101 Nicolls Road, Health Sciences Center Level 4, Stony Brook, New York, NY, 11794, USA.
| | - Brian M Zeglis
- Department of Chemistry, Hunter College of the City University of New York, New York, NY, USA.
- Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY, USA.
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiology, Weill Cornell Medical College, 413 East 69th Street, New York, NY, 10021, USA.
| |
Collapse
|
12
|
Rochet LNC, Bahou C, Wojciechowski JP, Koutsopetras I, Britton P, Spears RJ, Thanasi IA, Shao B, Zhong L, Bučar DK, Aliev AE, Porter MJ, Stevens MM, Baker JR, Chudasama V. Use of pyridazinediones for tuneable and reversible covalent cysteine modification applied to peptides, proteins and hydrogels. Chem Sci 2023; 14:13743-13754. [PMID: 38075666 PMCID: PMC10699563 DOI: 10.1039/d3sc04976k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 10/27/2023] [Indexed: 05/02/2024] Open
Abstract
Reversible cysteine modification has been found to be a useful tool for a plethora of applications such as selective enzymatic inhibition, activity-based protein profiling and/or cargo release from a protein or a material. However, only a limited number of reagents display reliable dynamic/reversible thiol modification and, in most cases, many of these reagents suffer from issues of stability, a lack of modularity and/or poor rate tunability. In this work, we demonstrate the potential of pyridazinediones as novel reversible and tuneable covalent cysteine modifiers. We show that the electrophilicity of pyridazinediones correlates to the rates of the Michael addition and retro-Michael deconjugation reactions, demonstrating that pyridazinediones provide an enticing platform for readily tuneable and reversible thiol addition/release. We explore the regioselectivity of the novel reaction and unveil the reason for the fundamental increased reactivity of aryl bearing pyridazinediones by using DFT calculations and corroborating findings with SCXRD. We also applied this fundamental discovery to making more rapid disulfide rebridging agents in related work. We finally provide the groundwork for potential applications in various areas with exemplification using readily functionalised "clickable" pyridazinediones on clinically relevant cysteine and disulfide conjugated proteins, as well as on a hydrogel material.
Collapse
Affiliation(s)
- Léa N C Rochet
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Calise Bahou
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Jonathan P Wojciechowski
- Department of Materials, Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London London SW7 2AZ UK
| | - Ilias Koutsopetras
- Bio-Functional Chemistry (UMR 7199), Institut du Médicament de Strasbourg, University of Strasbourg 74 Route du Rhin 67400 Illkirch-Graffenstaden France
| | - Phyllida Britton
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Richard J Spears
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Ioanna A Thanasi
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Baihao Shao
- Department of Materials, Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London London SW7 2AZ UK
| | - Lisha Zhong
- Department of Materials, Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London London SW7 2AZ UK
| | - Dejan-Krešimir Bučar
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Abil E Aliev
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Michael J Porter
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London London SW7 2AZ UK
| | - James R Baker
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Vijay Chudasama
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| |
Collapse
|
13
|
Delaney S, Nagy Á, Karlström AE, Zeglis BM. Site-Specific Photoaffinity Bioconjugation for the Creation of 89Zr-Labeled Radioimmunoconjugates. Mol Imaging Biol 2023; 25:1104-1114. [PMID: 37052759 PMCID: PMC10570397 DOI: 10.1007/s11307-023-01818-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/14/2023] [Accepted: 03/23/2023] [Indexed: 04/14/2023]
Abstract
PURPOSE Site-specific approaches to bioconjugation produce well-defined and homogeneous immunoconjugates with potential for superior in vivo behavior compared to analogs synthesized using traditional, stochastic methods. The possibility of incorporating photoaffinity chemistry into a site-specific bioconjugation strategy is particularly enticing, as it could simplify and accelerate the preparation of homogeneous immunoconjugates for the clinic. In this investigation, we report the synthesis, in vitro characterization, and in vivo evaluation of a site-specifically modified, 89Zr-labeled radioimmunoconjugate created via the reaction between an mAb and an Fc-binding protein bearing a photoactivatable 4-benzoylphenylalanine residue. PROCEDURES A variant of the Fc-binding Z domain of protein A containing a photoactivatable, 4-benzoylphenylalanine residue - Z(35BPA) - was modified with desferrioxamine (DFO), combined with the A33 antigen-targeting mAb huA33, and irradiated with UV light. The resulting immunoconjugate - DFOZ(35BPA)-huA33 - was purified and characterized via SDS-PAGE, MALDI-ToF mass spectrometry, surface plasmon resonance, and flow cytometry. The radiolabeling of DFOZ(35BPA)-huA33 was optimized to produce [89Zr]Zr-DFOZ(35BPA)-huA33, and the immunoreactivity of the radioimmunoconjugate was determined with SW1222 human colorectal cancer cells. Finally, the in vivo performance of [89Zr]Zr-DFOZ(35BPA)-huA33 in mice bearing subcutaneous SW1222 xenografts was interrogated via PET imaging and biodistribution experiments and compared to that of a stochastically labeled control radioimmunoconjugate, [89Zr]Zr-DFO-huA33. RESULTS HuA33 was site-specifically modified with Z(35BPA)-DFO, producing an immunoconjugate with on average 1 DFO/mAb, high in vitro stability, and high affinity for its target. [89Zr]Zr-DFOZ(35BPA)-huA33 was synthesized in 95% radiochemical yield and exhibited a specific activity of 2 mCi/mg and an immunoreactive fraction of ~ 0.85. PET imaging and biodistribution experiments revealed that high concentrations of the radioimmunoconjugate accumulated in tumor tissue (i.e., ~ 40%ID/g at 120 h p.i.) but also that the Z(35BPA)-bearing immunoPET probe produced higher uptake in the liver, spleen, and kidneys than its stochastically modified cousin, [89Zr]Zr-DFO-huA33. CONCLUSIONS Photoaffinity chemistry and an Fc-binding variant of the Z domain were successfully leveraged to create a novel site-specific strategy for the synthesis of radioimmunoconjugates. The probe synthesized using this method - DFOZ(35BPA)-huA33 - was well-defined and homogeneous, and the resulting radioimmunoconjugate ([89Zr]Zr-DFOZ(35BPA)-huA33) boasted high specific activity, stability, and immunoreactivity. While the site-specifically modified radioimmunoconjugate produced high activity concentrations in tumor tissue, it also yielded higher uptake in healthy organs than a stochastically modified analog, suggesting that optimization of this system is necessary prior to clinical translation.
Collapse
Affiliation(s)
- Samantha Delaney
- Department of Chemistry, Hunter College, City University of New York, New York, NY, USA
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ábel Nagy
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Amelie Eriksson Karlström
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden.
| | - Brian M Zeglis
- Department of Chemistry, Hunter College, City University of New York, New York, NY, USA.
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY, USA.
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY, USA.
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
14
|
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
|
15
|
Yue TC, Ge Y, Aprile FA, Ma MT, Pham TT, Long NJ. Site-Specific 68Ga Radiolabeling of Trastuzumab Fab via Methionine for ImmunoPET Imaging. Bioconjug Chem 2023; 34:1802-1810. [PMID: 37751398 PMCID: PMC10587866 DOI: 10.1021/acs.bioconjchem.3c00344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/10/2023] [Indexed: 09/28/2023]
Abstract
Bioconjugates of antibodies and their derivatives radiolabeled with β+-emitting radionuclides can be utilized for diagnostic PET imaging. Site-specific attachment of radioactive cargo to antibody delivery vectors provides homogeneous, well-defined immunoconjugates. Recent studies have demonstrated the utility of oxaziridine chemistry for site-specific labeling of methionine residues. Herein, we applied this approach to site-specifically radiolabel trastuzumab-derived Fab immunoconjugates with 68Ga, which can be used for in vivo PET imaging of HER2-positive breast cancer tumors. Initially, a reactive azide was introduced to a single solvent-accessible methionine residue in both the wild-type Fab and an engineered derivative containing methionine residue M74, utilizing the principles of oxaziridine chemistry. Subsequently, these conjugates were functionalized with a modified DFO chelator incorporating dibenzocyclooctyne. The resulting DFO-WT and DFO-M74 conjugates were radiolabeled with generator-produced [68Ga]Ga3+, to yield the novel PET radiotracers, [68Ga]Ga-DFO-WT and [68Ga]Ga-DFO-M74. In vitro and in vivo studies demonstrated that [68Ga]Ga-DFO-M74 exhibited a higher affinity for HER2 receptors. Biodistribution studies in mice bearing orthotopic HER2-positive breast tumors revealed a higher uptake of [68Ga]Ga-DFO-M74 in the tumor tissue, accompanied by rapid renal clearance, enabling clear delineation of tumors using PET imaging. Conversely, [68Ga]Ga-DFO-WT exhibited lower uptake and inferior image contrast compared to [68Ga]Ga-DFO-M74. Overall, the results demonstrate that the highly facile methionine-oxaziridine modification approach can be simply applied to the synthesis of stable and site-specifically modified radiolabeled antibody-chelator conjugates with favorable pharmacokinetics for PET imaging.
Collapse
Affiliation(s)
- Thomas
T. C. Yue
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, White City Campus, Wood Lane, London W120BZ, U.K.
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, Fourth Floor Lambeth Wing, St. Thomas’ Hospital, London SE17EH, U.K.
| | - Ying Ge
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, White City Campus, Wood Lane, London W120BZ, U.K.
| | - Francesco A. Aprile
- Department
of Chemistry and Institute of Chemical Biology, Molecular Sciences
Research Hub, Imperial College London, White City Campus, Wood Lane, London W120BZ, U.K.
| | - Michelle T. Ma
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, Fourth Floor Lambeth Wing, St. Thomas’ Hospital, London SE17EH, U.K.
| | - Truc T. Pham
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, Fourth Floor Lambeth Wing, St. Thomas’ Hospital, London SE17EH, U.K.
| | - Nicholas J. Long
- Department
of Chemistry and Institute of Chemical Biology, Molecular Sciences
Research Hub, Imperial College London, White City Campus, Wood Lane, London W120BZ, U.K.
| |
Collapse
|
16
|
Chen LA, Yu YH, Tian WT, Lin WC, Grauffel C, Wu CY, Chen CL, Lim C, Chu HM, Chang TW, Peng CJ. Site-specific Conjugation of 6 DOTA Chelators to a CA19-9-targeting scFv-Fc Antibody for Imaging and Therapy. J Med Chem 2023; 66:10604-10616. [PMID: 37462154 PMCID: PMC10424180 DOI: 10.1021/acs.jmedchem.3c00753] [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: 04/27/2023] [Indexed: 08/11/2023]
Abstract
Antibodies conjugated with diagnostic/therapeutic radionuclides are attractive options for inoperable cancers lacking accurate imaging methods and effective therapeutics, such as pancreatic cancer. Hence, we have produced an antibody radionuclide conjugate termed TE-1132 comprising a α-CA19-9 scFv-Fc that is site-specifically conjugated at each C-terminus to 3 DOTA chelators via a cysteine-containing peptide linker. The smaller scFv-Fc size facilitates diffusivity within solid tumors, whereas the chelator-to-antibody ratio of six enabled 177Lu-radiolabeled TE-1132 to exhibit high radioactivity up to 520 MBq/nmol. In mice bearing BxPC3 tumors, immuno-SPECT/CT imaging of [111In]In-TE-1132 and the biodistribution of [177Lu]Lu-TE-1132 showed selective tumor accumulation. Single and multiple doses of [177Lu]Lu-TE-1132 effectively inhibited the BxPC3 tumor growth and prolonged the survival of mice with no irreversible body weight loss or hematopoietic damage. The adequate pharmacokinetic parameters, prominent tumor accumulation, and efficacy with good safety in mice encourage the further investigation of theranostic TE-1132 for treating pancreatic cancer.
Collapse
Affiliation(s)
- Li-An Chen
- Immunwork,
Inc., Academia Rd., Sec.
1, Nangang, Taipei 11571, Taiwan
| | - Yueh-Hsiang Yu
- Immunwork,
Inc., Academia Rd., Sec.
1, Nangang, Taipei 11571, Taiwan
| | - Wei-Ting Tian
- Immunwork,
Inc., Academia Rd., Sec.
1, Nangang, Taipei 11571, Taiwan
| | - Wei-Chen Lin
- Immunwork,
Inc., Academia Rd., Sec.
1, Nangang, Taipei 11571, Taiwan
| | - Cédric Grauffel
- Immunwork,
Inc., Academia Rd., Sec.
1, Nangang, Taipei 11571, Taiwan
| | - Chun-Yi Wu
- Department
of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Beitou, Taipei 112, Taiwan
| | - Chuan-Lin Chen
- Department
of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Beitou, Taipei 112, Taiwan
| | - Carmay Lim
- Institute
of Biomedical Sciences, Academia Sinica, Academia Road, Taipei 115, Taiwan
| | - Hsing-Mao Chu
- Immunwork,
Inc., Academia Rd., Sec.
1, Nangang, Taipei 11571, Taiwan
| | - Tse-Wen Chang
- Immunwork,
Inc., Academia Rd., Sec.
1, Nangang, Taipei 11571, Taiwan
| | - Chi-Jiun Peng
- Immunwork,
Inc., Academia Rd., Sec.
1, Nangang, Taipei 11571, Taiwan
| |
Collapse
|
17
|
Rodriguez C, Delaney S, Sebastiano J, Sarrett SM, Cornejo MA, Thau S, Hosny MM, Zeglis BM. Site-selective radiolabeling using mushroom tyrosinase and the strain-promoted oxidation-controlled 1,2-quinone cycloaddition. RSC Adv 2023; 13:17705-17709. [PMID: 37313000 PMCID: PMC10258682 DOI: 10.1039/d3ra03486k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 06/15/2023] Open
Abstract
We report the in vitro characterization and in vivo evaluation of a novel 89Zr-labeled radioimmunoconjugate synthesized using a site-selective bioconjugation strategy based on the oxidation of tyrosinase residues exposed by the deglycosylation of the IgG and the subsequent strain-promoted oxidation-controlled 1,2-quinone cycloaddition between these amino acids and trans-cyclooctene-bearing cargoes. More specifically, we site-selectively modified a variant of the A33 antigen-targeting antibody huA33 with the chelator desferrioxamine (DFO), thereby producing an immunoconjugate (DFO-SPOCQhuA33) with equivalent antigen binding affinity to its parent immunoglobulin but attenuated affinity for the FcγRI receptor. This construct was subsequently radiolabeled with [89Zr]Zr4+ to create a radioimmunoconjugate - [89Zr]Zr-DFO-SPOCQhuA33 - in high yield and specific activity that exhibited excellent in vivo behavior in two murine models of human colorectal carcinoma.
Collapse
Affiliation(s)
- Cindy Rodriguez
- PhD Program in Chemistry, Graduate Center of the City University of New York New York New York 10016 USA
- Department of Chemistry, Hunter College, City University of New York New York New York 10065 USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center New York New York 10021 USA
| | - Samantha Delaney
- Department of Chemistry, Hunter College, City University of New York New York New York 10065 USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center New York New York 10021 USA
- PhD Program in Biochemistry, Graduate Center of the City University of New York New York New York 10016 USA
| | - Joni Sebastiano
- Department of Chemistry, Hunter College, City University of New York New York New York 10065 USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center New York New York 10021 USA
- PhD Program in Biochemistry, Graduate Center of the City University of New York New York New York 10016 USA
| | - Samantha M Sarrett
- Department of Chemistry, Hunter College, City University of New York New York New York 10065 USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center New York New York 10021 USA
- PhD Program in Biochemistry, Graduate Center of the City University of New York New York New York 10016 USA
| | - Mike A Cornejo
- PhD Program in Chemistry, Graduate Center of the City University of New York New York New York 10016 USA
- Department of Chemistry, Hunter College, City University of New York New York New York 10065 USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center New York New York 10021 USA
| | - Sarah Thau
- Department of Chemistry, Hunter College, City University of New York New York New York 10065 USA
| | - Meena M Hosny
- Department of Chemistry, Hunter College, City University of New York New York New York 10065 USA
| | - Brian M Zeglis
- PhD Program in Chemistry, Graduate Center of the City University of New York New York New York 10016 USA
- Department of Chemistry, Hunter College, City University of New York New York New York 10065 USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center New York New York 10021 USA
- PhD Program in Biochemistry, Graduate Center of the City University of New York New York New York 10016 USA
- Department of Radiology, Weill Cornell Medical College New York New York 10021 USA
| |
Collapse
|
18
|
Maiti R, Patel B, Patel N, Patel M, Patel A, Dhanesha N. Antibody drug conjugates as targeted cancer therapy: past development, present challenges and future opportunities. Arch Pharm Res 2023; 46:361-388. [PMID: 37071273 PMCID: PMC11345756 DOI: 10.1007/s12272-023-01447-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/26/2023] [Indexed: 04/19/2023]
Abstract
Antibody drug conjugates (ADCs) are promising cancer therapeutics with minimal toxicity as compared to small cytotoxic molecules alone and have shown the evidence to overcome resistance against tumor and prevent relapse of cancer. The ADC has a potential to change the paradigm of cancer chemotherapeutic treatment. At present, 13 ADCs have been approved by USFDA for the treatment of various types of solid tumor and haematological malignancies. This review covers the three structural components of an ADC-antibody, linker, and cytotoxic payload-along with their respective structure, chemistry, mechanism of action, and influence on the activity of ADCs. It covers comprehensive insight on structural role of linker towards efficacy, stability & toxicity of ADCs, different types of linkers & various conjugation techniques. A brief overview of various analytical techniques used for the qualitative and quantitative analysis of ADC is summarized. The current challenges of ADCs, such as heterogeneity, bystander effect, protein aggregation, inefficient internalization or poor penetration into tumor cells, narrow therapeutic index, emergence of resistance, etc., are outlined along with recent advances and future opportunities for the development of more promising next-generation ADCs.
Collapse
Affiliation(s)
- Ritwik Maiti
- Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India
| | - Bhumika Patel
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India.
| | - Nrupesh Patel
- Department of Pharmaceutical Analysis, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India
| | - Mehul Patel
- Department of Pharmaceutical Chemistry and Analysis, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT Campus, Changa, 388421, Gujarat, India
| | - Alkesh Patel
- Department of Pharmacology, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT Campus, Changa, 388421, Gujarat, India
| | - Nirav Dhanesha
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA.
| |
Collapse
|
19
|
Dewulf J, Flieswasser T, Delahaye T, Vangestel C, Miranda A, de Haard H, Jacobs J, Smits E, Van den Wyngaert T, Elvas F. Site-specific 68Ga-labeled nanobody for PET imaging of CD70 expression in preclinical tumor models. EJNMMI Radiopharm Chem 2023; 8:8. [PMID: 37093350 PMCID: PMC10126183 DOI: 10.1186/s41181-023-00194-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 04/20/2023] [Indexed: 04/25/2023] Open
Abstract
BACKGROUND CD70-CD27 is a costimulatory ligand-receptor pair in the tumor necrosis factor receptor family. With only limited expression in normal tissues, CD70 is constitutively expressed in a variety of solid tumors and hematologic malignancies, facilitating immunosuppression through CD27 signaling in the tumor microenvironment by enhanced survival of regulatory T cells, induction of T cell apoptosis, and T cell exhaustion. Consequently, CD70 is an increasingly recognized target for developing antibody-based therapies, but its expression patterns vary among different tumor types in spatial distribution, magnitude of expression and percentage of positive cells. In that regard, individual confirmation of CD70 expression at screening and during treatment could enhance the successful implementation of anti-CD70 therapies. Here, we developed a gallium-68 (68Ga) radiolabeled single-domain antibody-fragment targeting CD70 for in vivo positron emission tomography (PET) imaging. RESULTS An anti-CD70 VHH construct containing a C-direct-tag with a free thiol was developed to enable site-specific conjugation to a NOTA bifunctional chelator for 68Ga radiolabeling. [68Ga]Ga-NOTA-anti-CD70 VHH was obtained in good radiochemical yield of 30.4 ± 1.7% and high radiochemical purity (> 94%). The radiolabeled VHH showed excellent in vitro and in vivo stability. Specific binding of [68Ga]Ga-NOTA-anti-CD70 VHH was observed on CD70high 786-O cells, showing significantly higher cell-associated activity when compared to the blocking condition (p < 0.0001) and CD70low NCl-H1975 cells (p < 0.0001). PET imaging showed specific radiotracer accumulation in CD70 expressing human tumor xenografts, which was efficiently blocked by prior injection of unlabeled anti-CD70 VHH (p = 0.0029). In addition, radiotracer uptake in CD70high tumors was significantly higher when compared with CD70low tumors (p < 0.0001). The distribution of the radioactivity in the tumors using autoradiography was spatially matched with immunohistochemistry analysis of CD70 expression. CONCLUSION [68Ga]Ga-NOTA-anti-CD70 VHH showed excellent in vivo targeting of CD70 in human cancer xenografts. PET imaging using this radioimmunoconjugate holds promise as a non-invasive method to identify and longitudinally follow-up patients who will benefit most from anti-CD70 therapies.
Collapse
Affiliation(s)
- Jonatan Dewulf
- Molecular Imaging Center Antwerp (MICA), Integrated Personalized and Precision Oncology Network (IPPON), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Tal Flieswasser
- Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, IPPON, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Tim Delahaye
- argenx BV, Industriepark 7, Zwijnaarde, 9052, Gent, Belgium
| | - Christel Vangestel
- Molecular Imaging Center Antwerp (MICA), Integrated Personalized and Precision Oncology Network (IPPON), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
- Nuclear Medicine Department, Antwerp University Hospital, Drie Eikenstraat 655, 2650, Edegem, Belgium
| | - Alan Miranda
- Molecular Imaging Center Antwerp (MICA), Integrated Personalized and Precision Oncology Network (IPPON), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Hans de Haard
- argenx BV, Industriepark 7, Zwijnaarde, 9052, Gent, Belgium
| | - Julie Jacobs
- argenx BV, Industriepark 7, Zwijnaarde, 9052, Gent, Belgium
| | - Evelien Smits
- Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, IPPON, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Tim Van den Wyngaert
- Molecular Imaging Center Antwerp (MICA), Integrated Personalized and Precision Oncology Network (IPPON), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
- Nuclear Medicine Department, Antwerp University Hospital, Drie Eikenstraat 655, 2650, Edegem, Belgium
| | - Filipe Elvas
- Molecular Imaging Center Antwerp (MICA), Integrated Personalized and Precision Oncology Network (IPPON), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium.
| |
Collapse
|
20
|
Privat M, Bellaye PS, Chazeau E, Racoeur C, Adumeau P, Vivier D, Bernhard C, Moreau M, Collin B, Bettaieb A, Denat F, Bodio E, Paul C, Goze C. First Comparison Study of the In Vitro and In Vivo Properties of a Randomly and Site-Specifically Conjugated SPECT/NIRF Monomolecular Multimodal Imaging Probe (MOMIP) Based on an aza-BODIPY Fluorophore. Bioconjug Chem 2023. [DOI: 10.1021/acs.bioconjchem.3c00080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
21
|
Sapozhnikova KA, Gulyak EL, Brylev VA, Misyurin VA, Oreshkov SD, Alexeeva AV, Ryazantsev DY, Simonova MA, Ryabukhina EV, Popova GP, Tikhonova NA, Lyzhko NA, Barmashov AE, Misyurin AV, Ustinov AV, Alferova VA, Korshun VA. Aminooxy Click Modification of a Periodate-Oxidized Immunoglobulin G: A General Approach to Antibody-Drug Conjugates with Dye-Mediated Expeditious Stoichiometry Control. Int J Mol Sci 2023; 24:ijms24065134. [PMID: 36982208 PMCID: PMC10049567 DOI: 10.3390/ijms24065134] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/28/2023] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
A universal approach to the construction of antibody-drug conjugates (ADCs) has been developed. It relies on periodate oxidation of naturally present glycans of immunoglobulin G, followed by oxime ligation and, optionally, copper(I)-catalyzed alkyne-azide cycloaddition for conjugation with a toxic payload. The introduction of highly absorbing cyanine dyes into the linker allows for facile determination of the drug-antibody ratio. We applied this methodology to the synthesis of cytotoxic conjugates of an antibody against the tumor-associated antigen PRAME with doxorubicin and monomethyl auristatin E (MMAE). The resultant conjugates retained their affinity to a large extent, yet their cytotoxicity in vitro varied dramatically: while the doxorubicin-based conjugate did not produce any effect on cells, the MMAE-based one demonstrated specific activity against PRAME-expressing cancer cell lines. Importantly, the latter conjugate constitutes the first reported example of a PRAME-targeting ADC.
Collapse
Affiliation(s)
- Ksenia A Sapozhnikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Evgeny L Gulyak
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Vladimir A Brylev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
- Lumiprobe RUS Ltd., Kotsyubinskogo 4, 121351 Moscow, Russia
| | - Vsevolod A Misyurin
- N.N. Blokhin National Medical Cancer Research Center, Ministry of Health of Russia, Kashirskoye sh. 24, 115478 Moscow, Russia
| | - Sergey D Oreshkov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | | | - Dmitry Yu Ryazantsev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Maria A Simonova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Ekaterina V Ryabukhina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Galina P Popova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | | | | | - Alexander E Barmashov
- N.N. Blokhin National Medical Cancer Research Center, Ministry of Health of Russia, Kashirskoye sh. 24, 115478 Moscow, Russia
| | | | - Alexey V Ustinov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
- Lumiprobe RUS Ltd., Kotsyubinskogo 4, 121351 Moscow, Russia
| | - Vera A Alferova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Vladimir A Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| |
Collapse
|
22
|
Melendez-Alafort L, Ferro-Flores G, De Nardo L, Ocampo-García B, Bolzati C. Zirconium immune-complexes for PET molecular imaging: Current status and prospects. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.215005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
23
|
Maity S, Bingham C, Sheng W, Ehyaei N, Chakraborty D, Tahmasebi-Nick S, Kimmel TE, Vasileiou C, Geiger JH, Borhan B. Light controlled reversible Michael addition of cysteine: a new tool for dynamic site-specific labeling of proteins. Analyst 2023; 148:1085-1092. [PMID: 36722993 PMCID: PMC9992065 DOI: 10.1039/d2an01395a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Cysteine-based Michael addition is a widely employed strategy for covalent conjugation of proteins, peptides, and drugs. The covalent reaction is irreversible in most cases, leading to a lack of control over the process. Utilizing spectroscopic analyses along with X-ray crystallographic studies, we demonstrate Michael addition of an engineered cysteine residue in human Cellular Retinol Binding Protein II (hCRBPII) with a coumarin analog that creates a non-fluorescent complex. UV-illumination reverses the conjugation, yielding a fluorescent species, presumably through a retro-Michael process. This series of events can be repeated between a bound and non-bound form of the cysteine reversibly, resulting in the ON-OFF control of fluorescence. The details of the mechanism of photoswitching was illuminated by recapitulation of the process in light irradiated single crystals, confirming the mechanism at atomic resolution.
Collapse
Affiliation(s)
- Soham Maity
- Department of Chemistry, Michigan State University, 578 S. Shaw Ln., East Lansing, MI 48824, USA.
| | - Courtney Bingham
- Department of Chemistry, Michigan State University, 578 S. Shaw Ln., East Lansing, MI 48824, USA.
| | - Wei Sheng
- Roche Tissue Diagnostics, 1910 E Innovation Park Dr, Oro Valley, AZ, 85755, USA
| | - Nona Ehyaei
- Lycia Therapeutics, 400 East Jamie Court, S San Francisco, CA 94080, USA
| | - Debarshi Chakraborty
- Department of Chemistry, Michigan State University, 578 S. Shaw Ln., East Lansing, MI 48824, USA.
| | | | - Thomas E Kimmel
- Department of Chemistry, Michigan State University, 578 S. Shaw Ln., East Lansing, MI 48824, USA.
| | - Chrysoula Vasileiou
- Department of Chemistry, Michigan State University, 578 S. Shaw Ln., East Lansing, MI 48824, USA.
| | - James H Geiger
- Department of Chemistry, Michigan State University, 578 S. Shaw Ln., East Lansing, MI 48824, USA.
| | - Babak Borhan
- Department of Chemistry, Michigan State University, 578 S. Shaw Ln., East Lansing, MI 48824, USA.
| |
Collapse
|
24
|
Aigbogun OP, Phenix CP, Krol ES, Price EW. The Chemistry of Creating Chemically Programmed Antibodies (cPAbs): Site-Specific Bioconjugation of Small Molecules. Mol Pharm 2023; 20:853-874. [PMID: 36696533 DOI: 10.1021/acs.molpharmaceut.2c00821] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Small-molecule drugs have been employed for years as therapeutics in the pharmaceutical industry. However, small-molecule drugs typically have short in vivo half-lives which is one of the largest impediments to the success of many potentially valuable pharmacologically active small molecules. The undesirable pharmacokinetics and pharmacology associated with some small molecules have led to the development of a new class of bioconjugates known as chemically programmed antibodies (cPAbs). cPAbs are bioconjugates in which antibodies are used to augment small molecules with effector functions and prolonged pharmacokinetic profiles, where the pharmacophore of the small molecule is harnessed for target binding and therefore biological targeting. Many different small molecules can be conjugated to large proteins such as full monoclonal antibodies (IgG), fragment crystallizable regions (Fc), or fragment antigen binding regions (Fab). In order to successfully and site-specifically conjugate small molecules to any class of antibodies (IgG, Fc, or Fab), the molecules must be derivatized with a functional group for ease of conjugation without altering the pharmacology of the small molecules. In this Review, we summarize the different synthetic or biological methods that have been employed to produce cPAbs. These unique chemistries have potential to be applied to other fields of antibody modification such as antibody drug conjugates, radioimmunoconjugates, and fluorophore-tagged antibodies.
Collapse
Affiliation(s)
- Omozojie P Aigbogun
- Department of Chemistry, University of Saskatchewan, 110 Science Place, Saskatoon, S7N-5C9 Saskatchewan, Canada
| | - Christopher P Phenix
- Department of Chemistry, University of Saskatchewan, 110 Science Place, Saskatoon, S7N-5C9 Saskatchewan, Canada
| | - Ed S Krol
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, S7N-5E5 Saskatchewan, Canada
| | - Eric W Price
- Department of Chemistry, University of Saskatchewan, 110 Science Place, Saskatoon, S7N-5C9 Saskatchewan, Canada
| |
Collapse
|
25
|
Branched Linkers for Site-Specific Fluorescent Labeling of Antibodies. Molecules 2023; 28:molecules28010425. [PMID: 36615611 PMCID: PMC9822498 DOI: 10.3390/molecules28010425] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/28/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
Fluorescent antibodies have proved to be an invaluable tool for molecular biology and diagnostics. They are routinely produced by modification of lysine residues, which leads to high heterogeneity. As such, their affinity may be compromised if the antigen-binding site is affected, the probability of which increases along with the degree of labeling. In this work, we propose a methodology for the synthesis of site-specific antibody-dye conjugates with a high degree of labeling. To this end, we synthesized two oxyamine-based branched triazide linkers and coupled them with a periodate-oxidized anti-PRAME antibody 6H8; two oxyamine-based linear monoazide linkers of similar structure were used as controls. The azide-labeled antibodies were subsequently conjugated with fluorescent dyes via SPAAC, a copper-free click reaction. Compared to their counterparts made with linear linkers, the branched conjugates possessed a higher degree of labeling. The utility of the methodology was demonstrated in the detection of the PRAME protein on the surface of the cell by flow cytometry.
Collapse
|
26
|
Feng Y, Sarrett SM, Meshaw RL, Vaidyanathan G, Cornejo MA, Zeglis BM, Zalutsky MR. Site-Specific Radiohalogenation of a HER2-Targeted Single-Domain Antibody Fragment Using a Novel Residualizing Prosthetic Agent. J Med Chem 2022; 65:15358-15373. [PMID: 36368007 DOI: 10.1021/acs.jmedchem.2c01331] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Because of their rapid tumor accumulation and normal tissue clearance, single-domain antibody fragments (sdAbs) are an attractive vehicle for developing radiotherapeutics labeled with the α-emitter 211At. Herein, we have evaluated iso-[211At]AGMB-PODS, a prosthetic agent that combines a functionality for residualizing radiohalogens with a phenyloxadiazolyl methylsulfone (PODS) moiety for site-specific sdAb conjugation. Iso-[211At]AGMB-PODS and its radioiodinated analogue were evaluated for thiol-selective conjugation to anti-HER2 5F7 sdAb bearing a C-terminus GGC tail. Both radiohalogenated PODS-5F7GGC conjugates were synthesized in good radiochemical yields and retained high binding affinity on HER2-positive BT474 breast carcinoma cells. Iso-[211At]AGMB-PODS-5F7GGC was considerably more stable in vitro than its maleimide analogue in the presence of cysteine and human serum albumin (HSA) and exhibited excellent tumor uptake and high in vivo stability. Superior tumor-to-kidney activity ratios were seen for both radiohalogenated PODS-5F7GGC conjugates compared with [177Lu]Lu-DOTA-PODS-5F7GGC. These results suggest that iso-[211At]AGMB-PODS-5F7GGC warrants further evaluation for the treatment of HER2-expressing malignancies.
Collapse
Affiliation(s)
- Yutian Feng
- Department of Radiology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Samantha M. Sarrett
- Hunter College, City University of New York, New York, New York 10021, United States
- Ph.D. Programs in Biochemistry and Chemistry, The Graduate Center, City University of New York, New York, New York 10021, United States
| | - Rebecca L. Meshaw
- Department of Radiology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Ganesan Vaidyanathan
- Department of Radiology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Mike A. Cornejo
- Hunter College, City University of New York, New York, New York 10021, United States
- Ph.D. Programs in Biochemistry and Chemistry, The Graduate Center, City University of New York, New York, New York 10021, United States
| | - Brian M. Zeglis
- Hunter College, City University of New York, New York, New York 10021, United States
- Ph.D. Programs in Biochemistry and Chemistry, The Graduate Center, City University of New York, New York, New York 10021, United States
| | - Michael R. Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, North Carolina 27710, United States
| |
Collapse
|
27
|
Uzal-Varela R, Patinec V, Tripier R, Valencia L, Maneiro M, Canle M, Platas-Iglesias C, Esteban-Gómez D, Iglesias E. On the dissociation pathways of copper complexes relevant as PET imaging agents. J Inorg Biochem 2022; 236:111951. [PMID: 35963110 DOI: 10.1016/j.jinorgbio.2022.111951] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/15/2022] [Accepted: 07/27/2022] [Indexed: 02/07/2023]
Abstract
Several bifunctional chelators have been synthesized in the last years for the development of new 64Cu-based PET agents for in vivo imaging. When designing a metal-based PET probe, it is important to achieve high stability and kinetic inertness once the radioisotope is coordinated. Different competitive assays are commonly used to evaluate the possible dissociation mechanisms that may induce Cu(II) release in the body. Among them, acid-assisted dissociation tests or transchelation challenges employing EDTA or SOD are frequently used to evaluate both solution thermodynamics and the kinetic behavior of potential metal-based systems. Despite of this, the Cu(II)/Cu(I) bioreduction pathway that could be promoted by the presence of bioreductants still remains little explored. To fill this gap we present here a detailed spectroscopic study of the kinetic behavior of different macrocyclic Cu(II) complexes. The complexes investigated include the cross-bridge cyclam derivative [Cu(CB-TE1A)]+, whose structure was determined using single-crystal X-ray diffraction. The acid-assisted dissociation mechanism was investigated using HClO4 and HCl to analyse the effect of the counterion on the rate constants. The complexes were selected so that the effects of complex charge and coordination polyhedron could be assessed. Cyclic voltammetry experiments were conducted to investigate whether the reduction to Cu(I) falls within the window of common bioreducing agents. The most striking behavior concerns the [Cu(NO2Th)]2+ complex, a 1,4,7-triazacyclononane derivative containing two methylthiazolyl pendant arms. This complex is extremely inert with respect to dissociation following the acid-catalyzed mechanism, but dissociates rather quickly in the presence of a bioreductant like ascorbic acid.
Collapse
Affiliation(s)
- Rocío Uzal-Varela
- Centro de Investigacións Científicas Avanzadas (CICA) and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, 15071 A Coruña, Galicia, Spain
| | - Véronique Patinec
- Univ Brest, UMR-CNRS 6521 CEMCA, 6 avenue Victor le Gorgeu, 29238 Brest, France
| | - Raphaël Tripier
- Univ Brest, UMR-CNRS 6521 CEMCA, 6 avenue Victor le Gorgeu, 29238 Brest, France
| | - Laura Valencia
- Departamento de Química Inorgánica, Universidade de Vigo, Facultad de Ciencias, 36310 Pontevedra, Spain
| | - Marcelino Maneiro
- Departamento de Química Inorgánica, Universidade de Santiago de Compostela, Facultade de Ciencias, 27002 Lugo, Spain
| | - Moisés Canle
- Centro de Investigacións Científicas Avanzadas (CICA) and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, 15071 A Coruña, Galicia, Spain
| | - Carlos Platas-Iglesias
- Centro de Investigacións Científicas Avanzadas (CICA) and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, 15071 A Coruña, Galicia, Spain
| | - David Esteban-Gómez
- Centro de Investigacións Científicas Avanzadas (CICA) and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, 15071 A Coruña, Galicia, Spain.
| | - Emilia Iglesias
- Centro de Investigacións Científicas Avanzadas (CICA) and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, 15071 A Coruña, Galicia, Spain.
| |
Collapse
|
28
|
Sarrett SM, Rodriguez C, Rymarczyk G, Hosny MM, Keinänen O, Delaney S, Thau S, Krantz BA, Zeglis BM. Lysine-Directed Site-Selective Bioconjugation for the Creation of Radioimmunoconjugates. Bioconjug Chem 2022; 33:1750-1760. [PMID: 35946495 DOI: 10.1021/acs.bioconjchem.2c00354] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The synthesis of radioimmunoconjugates via the stochastic attachment of bifunctional chelators to lysines can yield heterogeneous products with suboptimal in vitro and in vivo behavior. In response to this, several site-selective approaches to bioconjugation have been developed, yet each has intrinsic drawbacks, such as the need for expensive reagents or the complexity of incorporating unnatural amino acids into IgGs. Herein, we describe the use of a simple and facile approach to lysine-directed site-selective bioconjugation for the generation of radioimmunoconjugates. This strategy relies upon on the selective modification of single lysine residues within each light chain of the monoclonal antibody (mAb) with a branched azide-bearing perfluorophenyl ester (PFP-bisN3) followed by the ligation of dibenzocyclooctyne (DBCO)-bearing payloads to these bioorthogonal handles via the strain-promoted azide-alkyne cycloaddition. This methodology was used to create [89Zr]Zr-SSKDFO-pertuzumab, a radioimmunoconjugate of the HER2-targeting mAb pertuzumab labeled with desferrioxamine (DFO) and the positron-emitting radiometal zirconium-89 (89Zr). [89Zr]Zr-SSKDFO-pertuzumab was compared to a pair of analogous probes: one synthesized via random lysine modification ([89Zr]Zr-DFO-pertuzumab) and another via thiol-maleimide chemistry ([89Zr]Zr-malDFO-pertuzumab). The bioconjugation strategy was assessed using ESI mass spectrometry, SDS-PAGE, and autoradiography. All three immunoconjugates demonstrated comparable binding to HER2 via flow cytometry and surface plasmon resonance (SPR), and 89Zr-labeled variants of each were synthesized in >99% radiochemical yield and molar activities of up to ∼55.5 GBq/μmol (10 mCi/mg). Subsequently, the in vivo behavior of this trio of 89Zr-immunoPET probes was interrogated in athymic nude mice bearing subcutaneous HER2-expressing BT-474 human breast cancer xenografts. [89Zr]Zr-SSKDFO-pertuzumab, [89Zr]Zr-malDFO-pertuzumab, and [89Zr]Zr-DFO-pertuzumab produced positron emission tomography (PET) images with high tumoral uptake and high tumor-to-healthy organ activity concentration ratios. A terminal biodistribution study complemented the PET results, revealing tumoral activity concentrations of 126.9 ± 50.3%ID/g, 86.9 ± 53.2%ID/g, and 92.5 ± 27.2%ID/g at 144 h post-injection for [89Zr]Zr-SSKDFO-pertuzumab, [89Zr]Zr-malDFO-pertuzumab, and [89Zr]Zr-DFO-pertuzumab, respectively. Taken together, the data clearly illustrate that this highly modular and facile approach to site-selective bioconjugation produces radioimmunoconjugates that are better-defined and more homogeneous than stochastically modified constructs and also exhibit excellent in vitro and in vivo performance. Furthermore, we contend that this lysine-directed strategy holds several key advantages over extant approaches to site-selective bioconjugation, especially in the context of production for the clinic.
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, 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 10021, 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 10021, United States.,Ph.D. Program in Chemistry, Graduate Center of the City University of New York, New York, New York 10016, United States
| | - Grzegorz Rymarczyk
- Advanced Proteome Therapeutics Inc, Boston, Massachusetts 02118, United States
| | - Meena M Hosny
- Department of Chemistry, Hunter College, City University of New York, New York, New York 10065, United States
| | - Outi 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 10021, United States.,Department of Chemistry, University of Helsinki, Helsinki, 00100, Finland
| | - Samantha Delaney
- Department of Chemistry, Hunter College, City University of New York, New York, New York 10065, United States.,Ph.D. Program in Biochemistry, 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 10021, United States
| | - Sarah Thau
- Department of Chemistry, Hunter College, City University of New York, New York, New York 10065, United States
| | - Benjamin A Krantz
- Advanced Proteome Therapeutics Inc, Boston, Massachusetts 02118, United States.,Department of Medicine, Division of Hematology and Medical Oncology, NYU Grossman School of Medicine, New York, New York 10016, 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, 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 10021, 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 Radiology, Weill Cornell Medical College, New York, New York 10021, United States
| |
Collapse
|
29
|
Luckau L, Groves K, Blencowe C, Scrimshaw S, Dent A, Quaglia M. Impact of Bioconjugation on Structure and Function of Antibodies for Use in Immunoassay by Hydrogen-Deuterium Exchange Mass Spectrometry. Front Mol Biosci 2022; 9:866843. [PMID: 35874615 PMCID: PMC9301968 DOI: 10.3389/fmolb.2022.866843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 06/15/2022] [Indexed: 12/02/2022] Open
Abstract
Monoclonal antibodies (mAbs) are widely used as analytical components in immunoassays to detect target molecules in applications such as clinical diagnostics, food analysis and drug discovery. Functional groups are often conjugated to lysine or cysteine residues to aid immobilization of mAbs or to enable their detection in an antibody antigen complex. Good assay performance depends on the affinity and specificity of the mAbs for the antigen. The conjugation reaction however can cause higher order structural (HOS) changes and ultimately affect the assay performance. In this study, four differently conjugated mAbs were selected as model systems and characterized by mass spectrometry. Particularly, intact protein analysis by liquid-chromatography mass-spectrometry (LC-MS) was performed to determine the amount and distribution of conjugation. Hydrogen deuterium exchange mass spectrometry (HDX-MS) experiments were carried out for the structural characterization of the conjugated mAbs. Immunoassay experiments were performed to monitor the effects of conjugation on the binding properties of the antibodies selected. Good agreement between the mass spectrometry and binding experiment results was found. Particularly, it was noted that the overall structural flexibility of the antibodies increases upon cysteine conjugation and decreases for lysine conjugation. The conjugation of mAbs with bulky functional groups tends to decrease the deuterium uptake kinetics due to induced steric effects. Overall, this study shows correlations between conjugation, structure and function of immunoassay antibodies and the benefits of mass spectrometry to improve understanding of the conjugation reaction and provide insights that can predict immunoassay performance.
Collapse
Affiliation(s)
- Luise Luckau
- National Measurement Laboratory at LGC, Teddington, United Kingdom
- *Correspondence: Luise Luckau,
| | - Kate Groves
- National Measurement Laboratory at LGC, Teddington, United Kingdom
| | - Chris Blencowe
- Fleet Bioprocessing Ltd., Hartley Wintney, United Kingdom
| | - Sam Scrimshaw
- Fleet Bioprocessing Ltd., Hartley Wintney, United Kingdom
| | - Alastair Dent
- Fleet Bioprocessing Ltd., Hartley Wintney, United Kingdom
| | - Milena Quaglia
- National Measurement Laboratory at LGC, Teddington, United Kingdom
| |
Collapse
|
30
|
Caers J, Duray E, Vrancken L, Marcion G, Bocuzzi V, De Veirman K, Krasniqi A, Lejeune M, Withofs N, Devoogdt N, Dumoulin M, Karlström AE, D’Huyvetter M. Radiotheranostic Agents in Hematological Malignancies. Front Immunol 2022; 13:911080. [PMID: 35865548 PMCID: PMC9294596 DOI: 10.3389/fimmu.2022.911080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/06/2022] [Indexed: 12/23/2022] Open
Abstract
Radioimmunotherapy (RIT) is a cancer treatment that combines radiation therapy with tumor-directed monoclonal antibodies (Abs). Although RIT had been introduced for the treatment of CD20 positive non-Hodgkin lymphoma decades ago, it never found a broad clinical application. In recent years, researchers have developed theranostic agents based on Ab fragments or small Ab mimetics such as peptides, affibodies or single-chain Abs with improved tumor-targeting capacities. Theranostics combine diagnostic and therapeutic capabilities into a single pharmaceutical agent; this dual application can be easily achieved after conjugation to radionuclides. The past decade has seen a trend to increased specificity, fastened pharmacokinetics, and personalized medicine. In this review, we discuss the different strategies introduced for the noninvasive detection and treatment of hematological malignancies by radiopharmaceuticals. We also discuss the future applications of these radiotheranostic agents.
Collapse
Affiliation(s)
- Jo Caers
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
- Department of Hematology, CHU de Liège, Liège, Belgium
- *Correspondence: Jo Caers,
| | - Elodie Duray
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
- Centre for Protein Engineering, Inbios, University of Liège, Liège, Belgium
| | - Louise Vrancken
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
- Department of Hematology, CHU de Liège, Liège, Belgium
| | - Guillaume Marcion
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
| | - Valentina Bocuzzi
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
| | - Kim De Veirman
- Department of Hematology and Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ahmet Krasniqi
- Laboratory of In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium
| | - Margaux Lejeune
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
| | - Nadia Withofs
- Department of Nuclear Medicine, CHU de Liège, Liège, Belgium
| | - Nick Devoogdt
- Laboratory of In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium
| | - Mireille Dumoulin
- Centre for Protein Engineering, Inbios, University of Liège, Liège, Belgium
| | - Amelie Eriksson Karlström
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Matthias D’Huyvetter
- Laboratory of In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
31
|
Yang E, Liu Q, Huang G, Liu J, Wei W. Engineering nanobodies for next-generation molecular imaging. Drug Discov Today 2022; 27:1622-1638. [PMID: 35331925 DOI: 10.1016/j.drudis.2022.03.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 02/04/2022] [Accepted: 03/17/2022] [Indexed: 12/12/2022]
Abstract
In recent years, nanobodies have emerged as ideal imaging agents for molecular imaging. Molecular nanobody imaging combines the specificity of nanobodies with the sensitivity of state-of-the-art molecular imaging modalities, such as positron emission tomography (PET). Given that modifications of nanobodies alter their pharmacokinetics (PK), the engineering strategies that combine nanobodies with radionuclides determine the effectiveness, reliability, and safety of the molecular imaging probes. In this review, we introduce conjugation strategies that have been applied to nanobodies, including random conjugation, 99mTc tricarbonyl chemistry, sortase A-mediated site-specific conjugation, maleimide-cysteine chemistry, and click chemistries. We also summarize the latest advances in nanobody tracers, emphasizing their preclinical and clinical use. In addition, we elaborate on nanobody-based near-infrared fluorescence (NIRF) imaging and image-guided surgery.
Collapse
Affiliation(s)
- Erpeng Yang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Qiufang Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China.
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China.
| |
Collapse
|
32
|
Adumeau P, Raavé R, Boswinkel M, Heskamp S, Wessels HJCT, van Gool AJ, Moreau M, Bernhard C, Da Costa L, Goncalves V, Denat F. Site-Specific, Platform-Based Conjugation Strategy for the Synthesis of Dual-Labeled Immunoconjugates for Bimodal PET/NIRF Imaging of HER2-Positive Tumors. Bioconjug Chem 2022; 33:530-540. [PMID: 35230093 DOI: 10.1021/acs.bioconjchem.2c00049] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Because positron emission tomography (PET) and optical imaging are very complementary, the combination of these two imaging modalities is very enticing in the oncology field. Such bimodal imaging generally relies on imaging agents bearing two different imaging reporters. In the bioconjugation field, this is mainly performed by successive random conjugations of the two reporters on the protein vector, but these random conjugations can alter the vector properties. In this study, we aimed at abrogating the heterogeneity of the bimodal imaging immunoconjugate and mitigating the impact of multiple random conjugations. A trivalent platform bearing a DFO chelator for 89Zr labeling, a NIR fluorophore, IRDye800CW, and a bioconjugation handle was synthesized. This bimodal probe was site-specifically grafted to trastuzumab via glycan engineering. This new bimodal immunoconjugate was then investigated in terms of radiochemistry, in vitro and in vivo, and compared to the clinically relevant random equivalent. In vitro and in vivo, our strategy provides several improvements over the current clinical standard. The combination of site-specific conjugation with the monomolecular platform reduced the heterogeneity of the final immunoconjugate, improved the resistance of the fluorophore toward radiobleaching, and reduced the nonspecific uptake in the spleen and liver compared to the standard random immunoconjugate. To conclude, the strategy developed is very promising for the synthesis of better defined dual-labeled immunoconjugates, although there is still room for improvement. Importantly, this conjugation strategy is highly modular and could be used for the synthesis of a wide range of dual-labeled immunoconjugates.
Collapse
Affiliation(s)
- Pierre Adumeau
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR 6302, CNRS, Université Bourgogne Franche-Comté, 9 Avenue Alain Savary, 21000 Dijon, France
| | - René Raavé
- Department of Medical Imaging, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
| | - Milou Boswinkel
- Department of Medical Imaging, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
| | - Hans J C T Wessels
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Centre, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Alain J van Gool
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Centre, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Mathieu Moreau
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR 6302, CNRS, Université Bourgogne Franche-Comté, 9 Avenue Alain Savary, 21000 Dijon, France
| | - Claire Bernhard
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR 6302, CNRS, Université Bourgogne Franche-Comté, 9 Avenue Alain Savary, 21000 Dijon, France
| | - Laurène Da Costa
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR 6302, CNRS, Université Bourgogne Franche-Comté, 9 Avenue Alain Savary, 21000 Dijon, France
| | - Victor Goncalves
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR 6302, CNRS, Université Bourgogne Franche-Comté, 9 Avenue Alain Savary, 21000 Dijon, France
| | - Franck Denat
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR 6302, CNRS, Université Bourgogne Franche-Comté, 9 Avenue Alain Savary, 21000 Dijon, France
| |
Collapse
|
33
|
Hagemans IM, Wierstra PJ, Steuten K, Molkenboer-Kuenen JDM, van Dalen D, Ter Beest M, van der Schoot JMS, Ilina O, Gotthardt M, Figdor CG, Scheeren FA, Heskamp S, Verdoes M. Multiscale imaging of therapeutic anti-PD-L1 antibody localization using molecularly defined imaging agents. J Nanobiotechnology 2022; 20:64. [PMID: 35109860 PMCID: PMC8811974 DOI: 10.1186/s12951-022-01272-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/17/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND While immune checkpoint inhibitors such as anti-PD-L1 antibodies have revolutionized cancer treatment, only subgroups of patients show durable responses. Insight in the relation between clinical response, PD-L1 expression and intratumoral localization of PD-L1 therapeutics could improve patient stratification. Therefore, we present the modular synthesis of multimodal antibody-based imaging tools for multiscale imaging of PD-L1 to study intratumoral distribution of PD-L1 therapeutics. RESULTS To introduce imaging modalities, a peptide containing a near-infrared dye (sulfo-Cy5), a chelator (DTPA), an azide, and a sortase-recognition motif was synthesized. This peptide and a non-fluorescent intermediate were used for site-specific functionalization of c-terminally sortaggable mouse IgG1 (mIgG1) and Fab anti-PD-L1. To increase the half-life of the Fab fragment, a 20 kDa PEG chain was attached via strain-promoted azide-alkyne cycloaddition (SPAAC). Biodistribution and imaging studies were performed with 111In-labeled constructs in 4T1 tumor-bearing mice. Comparing our site-specific antibody-conjugates with randomly conjugated antibodies, we found that antibody clone, isotype and method of DTPA conjugation did not change tumor uptake. Furthermore, addition of sulfo-Cy5 did not affect the biodistribution. PEGylated Fab fragment displayed a significantly longer half-life compared to unPEGylated Fab and demonstrated the highest overall tumor uptake of all constructs. PD-L1 in tumors was clearly visualized by SPECT/CT, as well as whole body fluorescence imaging. Immunohistochemistry staining of tumor sections demonstrated that PD-L1 co-localized with the fluorescent and autoradiographic signal. Intratumoral localization of the imaging agent could be determined with cellular resolution using fluorescent microscopy. CONCLUSIONS A set of molecularly defined multimodal antibody-based PD-L1 imaging agents were synthesized and validated for multiscale monitoring of PD-L1 expression and localization. Our modular approach for site-specific functionalization could easily be adapted to other targets.
Collapse
Affiliation(s)
- Iris M Hagemans
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| | - Peter J Wierstra
- Department of Medical Imaging, Nuclear Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kas Steuten
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| | - Janneke D M Molkenboer-Kuenen
- Department of Medical Imaging, Nuclear Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Duco van Dalen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| | - Martin Ter Beest
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johan M S van der Schoot
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Olga Ilina
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| | - Martin Gotthardt
- Department of Medical Imaging, Nuclear Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
- Division of Immunotherapy, Oncode Institute, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ferenc A Scheeren
- Department of Dermatology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Nuclear Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Martijn Verdoes
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
- Institute for Chemical Immunology, Nijmegen, The Netherlands.
| |
Collapse
|
34
|
Lee J, Kim J, Heo I, Kim SJ, Lee HY, Jang S, Jang KS, Yang CS, Lee Y, Yoo WC, Min SJ. One-Pot Bifunctionalization of Silica Nanoparticles Conjugated with Bioorthogonal Linkers: Application in Dual-modal Imaging. Biomater Sci 2022; 10:3540-3546. [DOI: 10.1039/d2bm00258b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Covalent surface modification of silica nanoparticles (SNPs) offers great potential for the development of multimodal nanomaterials for biomedical applications. Herein, we report the synthesis of covalently conjugated bifunctional SNPs and...
Collapse
|
35
|
Sapozhnikova KA, Misyurin VA, Ryazantsev DY, Kokin EA, Finashutina YP, Alexeeva AV, Ivanov IA, Kocharovskaya MV, Tikhonova NA, Popova GP, Alferova VA, Ustinov AV, Korshun VA, Brylev VA. Sensitive Immunofluorescent Detection of the PRAME Antigen Using a Practical Antibody Conjugation Approach. Int J Mol Sci 2021; 22:12845. [PMID: 34884647 PMCID: PMC8657778 DOI: 10.3390/ijms222312845] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 11/21/2021] [Accepted: 11/25/2021] [Indexed: 11/17/2022] Open
Abstract
Bioconjugation of antibodies with various payloads has diverse applications across various fields, including drug delivery and targeted imaging techniques. Fluorescent immunoconjugates provide a promising tool for cancer diagnostics due to their high brightness, specificity, stability and target affinity. Fluorescent antibodies are widely used in flow cytometry for fast and sensitive identification and collection of cells expressing the target surface antigen. Nonetheless, current approaches to fluorescent labeling of antibodies most often use random modification, along with a few rather sophisticated site-specific techniques. The aim of our work was to develop a procedure for fluorescent labeling of immunoglobulin G via periodate oxidation of antibody glycans, followed by oxime ligation with fluorescent oxyamines. Here, we report a novel technique based on an in situ oxime ligation of ethoxyethylidene-protected aminooxy compounds with oxidized antibody glycans. The approach is suitable for easy modification of any immunoglobulin G, while ensuring that antigen-binding domains remain intact, thus revealing various possibilities for fluorescent probe design. The technique was used to label an antibody to PRAME, a cancer-testis protein overexpressed in a number of cancers. A 6H8 monoclonal antibody to the PRAME protein was directly modified with protected-oxyamine derivatives of fluorescein-type dyes (FAM, Alexa488, BDP-FL); the stoichiometry of the resulting conjugates was characterized spectroscopically. The immunofluorescent conjugates obtained were applied to the analysis of bone marrow samples from patients with oncohematological diseases and demonstrated high efficiency in flow cytometry quantification. The approach can be applied for the development of various immunofluorescent probes for detection of diagnostic and prognostic markers, which can be useful in anticancer therapy.
Collapse
MESH Headings
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/immunology
- Antigens, Neoplasm/analysis
- Antigens, Neoplasm/immunology
- Bone Marrow/immunology
- Bone Marrow/metabolism
- Bone Marrow/pathology
- Cell Line, Tumor
- Fluorescent Antibody Technique/methods
- Fluorescent Dyes/chemistry
- Humans
- Immunoconjugates/chemistry
- Immunoconjugates/immunology
- Immunoconjugates/metabolism
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/metabolism
Collapse
Affiliation(s)
- Ksenia A. Sapozhnikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.S.); (D.Y.R.); (E.A.K.); (I.A.I.); (M.V.K.); (G.P.P.); (V.A.A.); (A.V.U.)
| | - Vsevolod A. Misyurin
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye Highway 23, 115478 Moscow, Russia; (V.A.M.); (Y.P.F.)
| | - Dmitry Y. Ryazantsev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.S.); (D.Y.R.); (E.A.K.); (I.A.I.); (M.V.K.); (G.P.P.); (V.A.A.); (A.V.U.)
| | - Egor A. Kokin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.S.); (D.Y.R.); (E.A.K.); (I.A.I.); (M.V.K.); (G.P.P.); (V.A.A.); (A.V.U.)
| | - Yulia P. Finashutina
- N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye Highway 23, 115478 Moscow, Russia; (V.A.M.); (Y.P.F.)
| | - Anastasiya V. Alexeeva
- Faculty of General Medicine, Pirogov Russian National Research Medical University, Ostrovityanova 1, 117992 Moscow, Russia;
| | - Igor A. Ivanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.S.); (D.Y.R.); (E.A.K.); (I.A.I.); (M.V.K.); (G.P.P.); (V.A.A.); (A.V.U.)
| | - Milita V. Kocharovskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.S.); (D.Y.R.); (E.A.K.); (I.A.I.); (M.V.K.); (G.P.P.); (V.A.A.); (A.V.U.)
- Moscow Institute of Physics and Technology, Institutsky Lane 9, 141700 Dolgoprudny, Russia
| | | | - Galina P. Popova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.S.); (D.Y.R.); (E.A.K.); (I.A.I.); (M.V.K.); (G.P.P.); (V.A.A.); (A.V.U.)
| | - Vera A. Alferova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.S.); (D.Y.R.); (E.A.K.); (I.A.I.); (M.V.K.); (G.P.P.); (V.A.A.); (A.V.U.)
- Gause Institute of New Antibiotics, B. Pirogovskaya 11, 119021 Moscow, Russia
| | - Alexey V. Ustinov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.S.); (D.Y.R.); (E.A.K.); (I.A.I.); (M.V.K.); (G.P.P.); (V.A.A.); (A.V.U.)
| | - Vladimir A. Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.S.); (D.Y.R.); (E.A.K.); (I.A.I.); (M.V.K.); (G.P.P.); (V.A.A.); (A.V.U.)
| | - Vladimir A. Brylev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (K.A.S.); (D.Y.R.); (E.A.K.); (I.A.I.); (M.V.K.); (G.P.P.); (V.A.A.); (A.V.U.)
| |
Collapse
|
36
|
Zhou Z, Meshaw R, Zalutsky MR, Vaidyanathan G. Site-Specific and Residualizing Linker for 18F Labeling with Enhanced Renal Clearance: Application to an Anti-HER2 Single-Domain Antibody Fragment. J Nucl Med 2021; 62:1624-1630. [PMID: 33637584 PMCID: PMC8612331 DOI: 10.2967/jnumed.120.261446] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 02/01/2021] [Indexed: 11/16/2022] Open
Abstract
Single-domain antibody fragments (sdAbs) are promising vectors for immuno-PET; however, better methods for labeling sdAbs with 18F are needed. Herein, we evaluate a site-specific strategy using an 18F residualizing motif and the anti-epidermal growth factor receptor 2 (HER2) sdAb 5F7 bearing an engineered C-terminal GGC tail (5F7GGC). Methods: 5F7GGC was site-specifically attached with a tetrazine-bearing agent via thiol-maleimide reaction. The resultant conjugate was labeled with 18F by inverse electron demand Diels-Alder cycloaddition with a trans-cyclooctene attached to 6-18F-fluoronicotinoyl moiety via a renal brush border enzyme-cleavable linker and a PEG4 chain (18F-5F7GGC). For comparisons, 5F7 sdAb was labeled using the prototypical residualizing agent, N-succinimidyl 3-(guanidinomethyl)-5-125I-iodobenzoate (iso-125I-SGMIB). The 2 labeled sdAbs were compared in paired-label studies performed in the HER2-expressing BT474M1 breast carcinoma cell line and athymic mice bearing BT474M1 subcutaneous xenografts. Small-animal PET/CT imaging after administration of 18F-5F7GGC in the above mouse model was also performed. Results:18F-5F7GGC was synthesized in an overall radiochemical yield of 8.9% ± 3.2% with retention of HER2 binding affinity and immunoreactivity. The total cell-associated and intracellular activity for 18F-5F7GGC was similar to that for coincubated iso-125I-SGMIB-5F7. Likewise, the uptake of 18F-5F7GGC in BT474M1 xenografts in mice was similar to that for iso-125I-SGMIB-5F7; however, 18F-5F7GGC exhibited significantly more rapid clearance from the kidney. Small-animal PET/CT imaging confirmed high uptake and retention in the tumor with very little background activity at 3 h except in the bladder. Conclusion: This site-specific and residualizing 18F-labeling strategy could facilitate clinical translation of 5F7 anti-HER2 sdAb as well as other sdAbs for immuno-PET.
Collapse
Affiliation(s)
- Zhengyuan Zhou
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | - Rebecca Meshaw
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | - Michael R Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | | |
Collapse
|
37
|
Jin Y, Liu B, Younis MH, Huang G, Liu J, Cai W, Wei W. Next-Generation Molecular Imaging of Thyroid Cancer. Cancers (Basel) 2021; 13:3188. [PMID: 34202358 PMCID: PMC8268517 DOI: 10.3390/cancers13133188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/20/2021] [Accepted: 06/22/2021] [Indexed: 12/15/2022] Open
Abstract
An essential aspect of thyroid cancer (TC) management is personalized and precision medicine. Functional imaging of TC with radioiodine and [18F]FDG has been frequently used in disease evaluation for several decades now. Recently, advances in molecular imaging have led to the development of novel tracers based on aptamer, peptide, antibody, nanobody, antibody fragment, and nanoparticle platforms. The emerging targets-including HER2, CD54, SHP2, CD33, and more-are promising targets for clinical translation soon. The significance of these tracers may be realized by outlining the way they support the management of TC. The provided examples focus on where preclinical investigations can be translated. Furthermore, advances in the molecular imaging of TC may inspire the development of novel therapeutic or theranostic tracers. In this review, we summarize TC-targeting probes which include transporter-based and immuno-based imaging moieties. We summarize the most recent evidence in this field and outline how these emerging strategies may potentially optimize clinical practice.
Collapse
Affiliation(s)
- Yuchen Jin
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
- Department of Nuclear Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 200233, China
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Beibei Liu
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People’s Hospital Affiliatede to Shanghai Jiao Tong University, Shanghai 200233, China;
| | - Muhsin H. Younis
- Departments of Radiology and Medical Physics, University of Wisconsin–Madison, Madison, WI 53705-2275, USA;
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin–Madison, Madison, WI 53705-2275, USA;
- Carbone Cancer Center, University of Wisconsin, Madison, WI 53705, USA
| | - Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
| |
Collapse
|
38
|
Bolzati C, Spolaore B. Enzymatic Methods for the Site-Specific Radiolabeling of Targeting Proteins. Molecules 2021; 26:3492. [PMID: 34201280 PMCID: PMC8229434 DOI: 10.3390/molecules26123492] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 12/19/2022] Open
Abstract
Site-specific conjugation of proteins is currently required to produce homogenous derivatives for medicine applications. Proteins derivatized at specific positions of the polypeptide chain can actually show higher stability, superior pharmacokinetics, and activity in vivo, as compared with conjugates modified at heterogeneous sites. Moreover, they can be better characterized regarding the composition of the derivatization sites as well as the conformational and activity properties. To this aim, several site-specific derivatization approaches have been developed. Among these, enzymes are powerful tools that efficiently allow the generation of homogenous protein-drug conjugates under physiological conditions, thus preserving their native structure and activity. This review will summarize the progress made over the last decade on the use of enzymatic-based methodologies for the production of site-specific labeled immunoconjugates of interest for nuclear medicine. Enzymes used in this field, including microbial transglutaminase, sortase, galactosyltransferase, and lipoic acid ligase, will be overviewed and their recent applications in the radiopharmaceutical field will be described. Since nuclear medicine can benefit greatly from the production of homogenous derivatives, we hope that this review will aid the use of enzymes for the development of better radio-conjugates for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Cristina Bolzati
- Institute of Condensed Matter Chemistry and Technologies for Energy ICMATE-CNR, Corso Stati Uniti, 4, I-35127 Padova, Italy
| | - Barbara Spolaore
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via Marzolo, 5, I-35131 Padova, Italy
- CRIBI Biotechnology Center, University of Padua, Viale G. Colombo, 3, I-35131 Padova, Italy
| |
Collapse
|
39
|
Chomet M, van Dongen GAMS, Vugts DJ. State of the Art in Radiolabeling of Antibodies with Common and Uncommon Radiometals for Preclinical and Clinical Immuno-PET. Bioconjug Chem 2021; 32:1315-1330. [PMID: 33974403 PMCID: PMC8299458 DOI: 10.1021/acs.bioconjchem.1c00136] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
Inert
and stable radiolabeling of monoclonal antibodies (mAb),
antibody fragments, or antibody mimetics with radiometals is a prerequisite
for immuno-PET. While radiolabeling is preferably fast, mild, efficient,
and reproducible, especially when applied for human use in a current
Good Manufacturing Practice compliant way, it is crucial that the
obtained radioimmunoconjugate is stable and shows preserved immunoreactivity
and in vivo behavior. Radiometals and chelators have
extensively been evaluated to come to the most ideal radiometal–chelator
pair for each type of antibody derivative. Although PET imaging of
antibodies is a relatively recent tool, applications with 89Zr, 64Cu, and 68Ga have greatly increased in
recent years, especially in the clinical setting, while other less
common radionuclides such as 52Mn, 86Y, 66Ga, and 44Sc, but also 18F as in [18F]AlF are emerging promising candidates for the radiolabeling
of antibodies. This review presents a state of the art overview of
the practical aspects of radiolabeling of antibodies, ranging from
fast kinetic affibodies and nanobodies to slow kinetic intact mAbs.
Herein, we focus on the most common approach which consists of first
modification of the antibody with a chelator, and after eventual storage
of the premodified molecule, radiolabeling as a second step. Other
approaches are possible but have been excluded from this review. The
review includes recent and representative examples from the literature
highlighting which radiometal–chelator–antibody combinations
are the most successful for in vivo application.
Collapse
Affiliation(s)
- Marion Chomet
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam 1081 HV, The Netherlands
| | - Guus A M S van Dongen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam 1081 HV, The Netherlands
| | - Danielle J Vugts
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam 1081 HV, The Netherlands
| |
Collapse
|
40
|
Sharma SK, Adumeau P, Keinänen O, Sisodiya V, Sarvaiya H, Tchelepi R, Korsen JA, Pourat J, Edwards KJ, Ragupathi A, Hamdy O, Saunders LR, Rudin CM, Poirier JT, Lewis JS, Zeglis BM. Synthesis and Comparative In Vivo Evaluation of Site-Specifically Labeled Radioimmunoconjugates for DLL3-Targeted ImmunoPET. Bioconjug Chem 2021; 32:1255-1262. [PMID: 33835770 PMCID: PMC8295218 DOI: 10.1021/acs.bioconjchem.1c00121] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Delta-like ligand 3 (DLL3) is a therapeutic target for the treatment of small cell lung cancer, neuroendocrine prostate cancer, and isocitrate dehydrogenase mutant glioma. In the clinic, DLL3-targeted 89Zr-immunoPET has the potential to aid in the assessment of disease burden and facilitate the selection of patients suitable for therapies that target the antigen. The overwhelming majority of 89Zr-labeled radioimmunoconjugates are synthesized via the random conjugation of desferrioxamine (DFO) to lysine residues within the immunoglobulin. While this approach is admittedly facile, it can produce heterogeneous constructs with suboptimal in vitro and in vivo behavior. In an effort to circumvent these issues, we report the development and preclinical evaluation of site-specifically labeled radioimmunoconjugates for DLL3-targeted immunoPET. To this end, we modified a cysteine-engineered variant of the DLL3-targeting antibody SC16-MB1 with two thiol-reactive variants of DFO: one bearing a maleimide moiety (Mal-DFO) and the other containing a phenyloxadiazolyl methyl sulfone group (PODS-DFO). In an effort to obtain immunoconjugates with a DFO-to-antibody ratio (DAR) of 2, we explored both the reduction of the antibody with tris(2-carboxyethyl) phosphine (TCEP) as well as the use of a combination of glutathione and arginine as reducing and stabilizing agents, respectively. While exerting control over the DAR of the immunoconjugate proved cumbersome using TCEP, the use of glutathione and arginine enabled the selective reduction of the engineered cysteines and thus the formation of homogeneous immunoconjugates. A head-to-head comparison of the resulting 89Zr-radioimmunoconjugates in mice bearing DLL3-expressing H82 xenografts revealed no significant differences in tumoral uptake and showed comparable radioactivity concentrations in most healthy nontarget organs. However, 89Zr-DFOPODS-DAR2SC16-MB1 produced 30% lower uptake (3.3 ± 0.5 %ID/g) in the kidneys compared to 89Zr-DFOMal-DAR2SC16-MB1 (4.7 ± 0.5 %ID/g). In addition, H82-bearing mice injected with a 89Zr-labeled isotype-control radioimmunoconjugate synthesized using PODS exhibited ∼40% lower radioactivity in the kidneys compared to mice administered its maleimide-based counterpart. Taken together, these results demonstrate the improved in vivo performance of the PODS-based radioimmunoconjugate and suggest that a stable, well-defined DAR2 radiopharmaceutical may be suitable for the clinical immunoPET of DLL3-expressing cancers.
Collapse
Affiliation(s)
- Sai Kiran Sharma
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Department of Chemistry, Hunter College, City University of New York, New York, New York 10021, United States
| | - Pierre Adumeau
- Department of Chemistry, Hunter College, City University of New York, New York, New York 10021, United States
| | - Outi Keinänen
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Department of Chemistry, Hunter College, City University of New York, New York, New York 10021, United States
| | - Vikram Sisodiya
- Abbvie Stemcentrx, South San Francisco, California 94080, United States
| | - Hetal Sarvaiya
- Abbvie Stemcentrx, South San Francisco, California 94080, United States
| | - Robert Tchelepi
- Abbvie Stemcentrx, South San Francisco, California 94080, United States
| | - Joshua A Korsen
- Department of Pharmacology, Weill Cornell Medical College, New York, New York 10021, United States
| | - Jacob Pourat
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Kimberly J Edwards
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Ashwin Ragupathi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Omar Hamdy
- Abbvie Stemcentrx, South San Francisco, California 94080, United States
| | - Laura R Saunders
- Abbvie Stemcentrx, South San Francisco, California 94080, United States
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - John T Poirier
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Molecular Pharmacology Program, 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.,Department of Radiology, Weill Cornell Medical College, New York, New York 10021, United States.,Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Brian M Zeglis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Department of Chemistry, Hunter College, City University of New York, New York, New York 10021, United States.,Department of Radiology, Weill Cornell Medical College, New York, New York 10021, United States
| |
Collapse
|
41
|
Klika KD, Da Pieve C, Kopka K, Smith G, Makarem A. Synthesis and application of a thiol-reactive HBED-type chelator for development of easy-to-produce Ga-radiopharmaceutical kits and imaging probes. Org Biomol Chem 2021; 19:1722-1726. [PMID: 33527964 DOI: 10.1039/d0ob02513e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In radiopharmaceutical syntheses, maleimide is commonly used for linking thiol-bearing bioactive molecules to metal-complexing ligands (chelators). However, due to instability of the resulting linkage, phenyloxadiazolyl methylsulfone (PODS) was developed as an alternative to maleimide. This coupling strategy has never been attempted with HBED which is a powerful chelator for gallium-radiolabeling especially at ambient temperature. Here we present HBED-CC-PODS as a bifunctional chelator scaffold for the site-selective conjugation of thiol-bearing vectors and [68Ga]Ga-radiolabeling.
Collapse
Affiliation(s)
- Karel D Klika
- German Cancer Research Center (DKFZ), Molecular Structure Analysis, INF 280, 69120 Heidelberg, Germany
| | - Chiara Da Pieve
- The Institute of Cancer Research, Division of Radiotherapy and Imaging, 123 Old Brompton Road, London SW7 3RP, UK
| | - Klaus Kopka
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Graham Smith
- The Institute of Cancer Research, Division of Radiotherapy and Imaging, 123 Old Brompton Road, London SW7 3RP, UK
| | - Ata Makarem
- German Cancer Research Center (DKFZ), Division of Radiopharmaceutical Chemistry, INF 280, 69120 Heidelberg, Germany
| |
Collapse
|
42
|
Microfluidic Preparation of 89Zr-Radiolabelled Proteins by Flow Photochemistry. Molecules 2021; 26:molecules26030764. [PMID: 33540712 PMCID: PMC7867232 DOI: 10.3390/molecules26030764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 02/03/2023] Open
Abstract
89Zr-radiolabelled proteins functionalised with desferrioxamine B are a cornerstone of diagnostic positron emission tomography. In the clinical setting, 89Zr-labelled proteins are produced manually. Here, we explore the potential of using a microfluidic photochemical flow reactor to prepare 89Zr-radiolabelled proteins. The light-induced functionalisation and 89Zr-radiolabelling of human serum albumin ([89Zr]ZrDFO-PEG3-Et-azepin-HSA) was achieved by flow photochemistry with a decay-corrected radiochemical yield (RCY) of 31.2 ± 1.3% (n = 3) and radiochemical purity >90%. In comparison, a manual batch photoreactor synthesis produced the same radiotracer in a decay-corrected RCY of 59.6 ± 3.6% (n = 3) with an equivalent RCP > 90%. The results indicate that photoradiolabelling in flow is a feasible platform for the automated production of protein-based 89Zr-radiotracers, but further refinement of the apparatus and optimisation of the method are required before the flow process is competitive with manual reactions.
Collapse
|
43
|
Hentrich C, Kellmann SJ, Putyrski M, Cavada M, Hanuschka H, Knappik A, Ylera F. Periplasmic expression of SpyTagged antibody fragments enables rapid modular antibody assembly. Cell Chem Biol 2021; 28:813-824.e6. [PMID: 33529581 DOI: 10.1016/j.chembiol.2021.01.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/16/2020] [Accepted: 01/06/2021] [Indexed: 12/15/2022]
Abstract
Antibodies are essential tools in research and diagnostics. Although antibody fragments typically obtained from in vitro selection can be rapidly produced in bacteria, the generation of full-length antibodies or the modification of antibodies with probes is time and labor intensive. Protein ligation such as SpyTag technology could covalently attach domains and labels to antibody fragments equipped with a SpyTag. However, we found that the established periplasmic expression of antibody fragments in E. coli led to quantitative cleavage of the SpyTag by the proteases Tsp and OmpT. Here we report successful periplasmic expression of SpyTagged Fab fragments and demonstrate the coupling to separately prepared SpyCatcher modules. We used this modular toolbox of SpyCatcher proteins to generate reagents for a variety of immunoassays and measured their performance in comparison with traditional reagents. Furthermore, we demonstrate surface immobilization, high-throughput screening of antibody libraries, and rapid prototyping of antibodies based on modular antibody assembly.
Collapse
Affiliation(s)
| | | | - Mateusz Putyrski
- Bio-Rad AbD Serotec GmbH, Zeppelinstraße 4, 82178 Puchheim, Germany
| | - Manuel Cavada
- Bio-Rad AbD Serotec GmbH, Zeppelinstraße 4, 82178 Puchheim, Germany
| | - Hanh Hanuschka
- Bio-Rad AbD Serotec GmbH, Zeppelinstraße 4, 82178 Puchheim, Germany
| | - Achim Knappik
- Bio-Rad AbD Serotec GmbH, Zeppelinstraße 4, 82178 Puchheim, Germany
| | - Francisco Ylera
- Bio-Rad AbD Serotec GmbH, Zeppelinstraße 4, 82178 Puchheim, Germany.
| |
Collapse
|
44
|
Mukai H, Watanabe Y. Review: PET imaging with macro- and middle-sized molecular probes. Nucl Med Biol 2021; 92:156-170. [PMID: 32660789 DOI: 10.1016/j.nucmedbio.2020.06.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 12/16/2022]
Abstract
Recent progress in radiolabeling of macro- and middle-sized molecular probes has been extending possibilities to use PET molecular imaging for dynamic application to drug development and therapeutic evaluation. Theranostics concept also accelerated the use of macro- and middle-sized molecular probes for sharpening the contrast of proper target recognition even the cellular types/subtypes and proper selection of the patients who should be treated by the same molecules recognition. Here, brief summary of the present status of immuno-PET, and then further development of advanced technologies related to immuno-PET, peptidic PET probes, and nucleic acids PET probes are described.
Collapse
Affiliation(s)
- Hidefumi Mukai
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| |
Collapse
|
45
|
Wu AM. Protein Engineering for Molecular Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00045-4] [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
|
46
|
Antibody-Based Molecular Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00024-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
47
|
Feng Y, Zhou Z, McDougald D, Meshaw RL, Vaidyanathan G, Zalutsky MR. Site-specific radioiodination of an anti-HER2 single domain antibody fragment with a residualizing prosthetic agent. Nucl Med Biol 2021; 92:171-183. [PMID: 32448731 PMCID: PMC7657985 DOI: 10.1016/j.nucmedbio.2020.05.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 11/28/2022]
Abstract
INTRODUCTION As a consequence of their small size, high stability and high affinity, single domain antibody fragments (sdAbs) are appealing targeting vectors for radiopharmaceutical development. With sdAbs binding to internalizing receptors like HER2, residualizing prosthetic agents can enhance tumor retention of radioiodine, which until now has been done with random labeling approaches. Herein we evaluate a site-specific strategy utilizing a radioiodinated, residualizing maleimido moiety and the anti-HER2 sdAb 5F7 bearing a GGC tail for conjugation. METHODS Maleimidoethyl 3-(guanidinomethyl)-5-iodobenzoate ([131I]MEGMB) and its N-succinimidyl ester analogue, iso-[125I]SGMIB, were labeled by halodestannylation and conjugated with 5F7GGC and 5F7, respectively. Radiochemical purity, immunoreactivity and binding affinity were determined. Paired-label experiments directly compared iso-[125I]SGMIB-5F7 and [131I]MEGMIB-5F7GGC with regard to internalization/residualization and affinity on HER2-expressing SKOV-3 ovarian carcinoma cells as well as biodistribution and metabolite distribution in athymic mice with subcutaneous SKOV-3 xenografts. RESULTS [131I]MEGMIB-5F7GGC had an immunoreactivity of 81.3% and Kd = 0.94 ± 0.27 nM. Internalization assays demonstrated high intracellular trapping for both conjugates, For example, at 1 h, intracellular retention was 50.30 ± 3.36% for [131I]MEGMIB-5F7GGC and 55.95 ± 3.27% for iso-[125I]SGMIB-5F7, while higher retention was seen for iso-[125I]SGMIB-5F7 at later time points. Peak tumor uptake was similar for both conjugates (8.35 ± 2.66%ID/g and 8.43 ± 2.84%ID/g for iso-[125I]SGMIB-5F7 and [131I]MEGMIB-5F7GGC at 1 h, respectively); however, more rapid normal tissue clearance was seen for [131I]MEGMIB-5F7GGC, with a 2-fold higher tumor-to-kidney ratio and a 3-fold higher tumor-to-liver ratio compared with co-injected iso-[125I]SGMIB-5F7. Consisted with this, generation of labeled catabolites in the kidneys was higher for [131I]MEGMIB-5F7GGC. CONCLUSION [131I]MEGMIB-5F7GGC offers similar tumor targeting as iso-[125I]SGMIB-5F7 but with generally lower normal tissue uptake. ADVANCES IN KNOWLEDGE AND IMPLICATION FOR PATIENT CARE The site specific nature of the [131I]MEGMIB reagent may facilitate clinical translation, particularly for sdAb with compromised affinity after random labeling.
Collapse
Affiliation(s)
- Yutian Feng
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Zhengyuan Zhou
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Darryl McDougald
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Rebecca L Meshaw
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | | | - Michael R Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
48
|
Khozeimeh Sarbisheh E, Dewaele-Le Roi G, Shannon WE, Tan S, Xu Y, Zeglis BM, Price EW. DiPODS: A Reagent for Site-Specific Bioconjugation via the Irreversible Rebridging of Disulfide Linkages. Bioconjug Chem 2020; 31:2789-2806. [PMID: 33210532 DOI: 10.1021/acs.bioconjchem.0c00590] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Chemoselective reactions with thiols have long held promise for the site-specific bioconjugation of antibodies and antibody fragments. Yet bifunctional probes bearing monovalent maleimides-long the "gold standard" for thiol-based ligations-are hampered by two intrinsic issues: the in vivo instability of the maleimide-thiol bond and the need to permanently disrupt disulfide linkages in order to facilitate bioconjugation. Herein, we present the synthesis, characterization, and validation of DiPODS, a novel bioconjugation reagent containing a pair of oxadiazolyl methyl sulfone moieties capable of irreversibly forming covalent bonds with two thiolate groups while simultaneously rebridging disulfide linkages. The reagent was synthesized from commercially available starting materials in 8 steps, during which rotamers were encountered and investigated both experimentally and computationally. DiPODS is designed to be modular and can thus be conjugated to any payload through a pendant terminal primary amine (DiPODS-PEG4-NH2). Subsequently, the modification of a HER2-targeting Fab with a fluorescein-conjugated variant of DiPODS (DiPODS-PEG4-FITC) reinforced the site-specificity of the reagent, illustrated its ability to rebridge disulfide linkages, and produced an immunoconjugate with in vitro properties superior to those of an analogous construct created using traditional stochastic bioconjugation techniques. Ultimately, we believe that this work has particularly important implications for the synthesis of immunoconjugates, specifically for ensuring that the attachment of cargoes to immunoglobulins is robust, irreversible, and biologically and structurally benign.
Collapse
Affiliation(s)
| | - Guillaume Dewaele-Le Roi
- 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 10021, United States.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Whitney E Shannon
- Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N-5C9, Canada
| | - Sally Tan
- Department of Chemistry, Hunter College, City University of New York, New York, New York 10021, United States
| | - Yujia Xu
- 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 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 10021, United States.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Eric W Price
- Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N-5C9, Canada
| |
Collapse
|
49
|
Light-induced synthesis of protein conjugates and its application in photoradiosynthesis of 89Zr-radiolabeled monoclonal antibodies. Nat Protoc 2020; 15:3579-3594. [DOI: 10.1038/s41596-020-0386-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/22/2020] [Indexed: 12/31/2022]
|
50
|
Wall A, Wills AG, Forte N, Bahou C, Bonin L, Nicholls K, Ma MT, Chudasama V, Baker JR. One-pot thiol-amine bioconjugation to maleimides: simultaneous stabilisation and dual functionalisation. Chem Sci 2020; 11:11455-11460. [PMID: 34094388 PMCID: PMC8162801 DOI: 10.1039/d0sc05128d] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 09/23/2020] [Indexed: 11/21/2022] Open
Abstract
Maleimide chemistry is widely used in the site-selective modification of proteins. However, hydrolysis of the resultant thiosuccinimides is required to provide robust stability to the bioconjugates. Herein, we present an alternative approach that affords simultaneous stabilisation and dual functionalisation in a one pot fashion. By consecutive conjugation of a thiol and an amine to dibromomaleimides, we show that aminothiomaleimides can be generated extremely efficiently. Furthermore, the amine serves to deactivate the electrophilicity of the maleimide, precluding further reactivity and hence generating stable conjugates. We have applied this conjugation strategy to peptides and proteins to generate stabilised trifunctional conjugates. We propose that this stabilisation-dual modification strategy could have widespread use in the generation of diverse conjugates.
Collapse
Affiliation(s)
- Archie Wall
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Alfie G Wills
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Nafsika Forte
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Calise Bahou
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Lisa Bonin
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | | | - Michelle T Ma
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London SE1 7EH UK
| | - Vijay Chudasama
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa Lisbon Portugal
| | - James R Baker
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| |
Collapse
|