1
|
Liu S, Li Z, Huisman M, Shah DK. Clinical validation of translational antibody PBPK model using tissue distribution data generated with 89Zr-immuno-PET imaging. J Pharmacokinet Pharmacodyn 2023; 50:377-394. [PMID: 37382712 DOI: 10.1007/s10928-023-09869-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/09/2023] [Indexed: 06/30/2023]
Abstract
The main objective of this manuscript was to validate the ability of the monoclonal antibody physiologically-based pharmacokinetic (PBPK) model to predict tissue concentrations of antibodies in the human. To accomplish this goal, preclinical and clinical tissue distribution and positron emission tomography imaging data generated using zirconium-89 (89Zr) labeled antibodies were obtained from the literature. First, our previously published translational PBPK model for antibodies was expanded to describe the whole-body biodistribution of 89Zr labeled antibody and the free 89Zr, as well as residualization of free 89Zr. Subsequently, the model was optimized using mouse biodistribution data, where it was observed that free 89Zr mainly residualizes in the bone and the extent of antibody distribution in certain tissues (e.g., liver and spleen) may be altered by labeling with 89Zr. The mouse PBPK model was scaled to rat, monkey, and human by simply changing the physiological parameters, and a priori simulations performed by the model were compared with the observed PK data. It was found that model predicted antibody PK in majority of the tissues in all the species superimposed over the observed data, and the model was also able to predict the PK of antibody in human tissues reasonably well. As such, the work presented here provides unprecedented evaluation of the antibody PPBK model for its ability to predict tissue PK of antibodies in the clinic. This model can be used for preclinical-to-clinical translation of antibodies and for prediction of antibody concentrations at the site-of-action in the clinic.
Collapse
Affiliation(s)
- Shufang Liu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, NY, 14214-8033, USA
| | - Zhe Li
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, NY, 14214-8033, USA
| | - Marc Huisman
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, NY, 14214-8033, USA.
| |
Collapse
|
2
|
ImmunoPET Directed to the Brain: A New Tool for Preclinical and Clinical Neuroscience. Biomolecules 2023; 13:biom13010164. [PMID: 36671549 PMCID: PMC9855881 DOI: 10.3390/biom13010164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/08/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Immuno-positron emission tomography (immunoPET) is a non-invasive in vivo imaging method based on tracking and quantifying radiolabeled monoclonal antibodies (mAbs) and other related molecules, such as antibody fragments, nanobodies, or affibodies. However, the success of immunoPET in neuroimaging is limited because intact antibodies cannot penetrate the blood-brain barrier (BBB). In neuro-oncology, immunoPET has been successfully applied to brain tumors because of the compromised BBB. Different strategies, such as changes in antibody properties, use of physiological mechanisms in the BBB, or induced changes to BBB permeability, have been developed to deliver antibodies to the brain. These approaches have recently started to be applied in preclinical central nervous system PET studies. Therefore, immunoPET could be a new approach for developing more specific PET probes directed to different brain targets.
Collapse
|
3
|
Syvänen S, Meier SR, Roshanbin S, Xiong M, Faresjö R, Gustavsson T, Bonvicini G, Schlein E, Aguilar X, Julku U, Eriksson J, Sehlin D. PET Imaging in Preclinical Anti-Aβ Drug Development. Pharm Res 2022; 39:1481-1496. [PMID: 35501533 PMCID: PMC9246809 DOI: 10.1007/s11095-022-03277-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/25/2022] [Indexed: 11/21/2022]
Abstract
Positron emission tomography (PET), a medical imaging technique allowing for studies of the living human brain, has gained an important role in clinical trials of novel drugs against Alzheimer’s disease (AD). For example, PET data contributed to the conditional approval in 2021 of aducanumab, an antibody directed towards amyloid-beta (Aβ) aggregates, by showing a dose-dependent reduction in brain amyloid after treatment. In parallel to clinical studies, preclinical studies in animal models of Aβ pathology may also benefit from PET as a tool to detect target engagement and treatment effects of anti-Aβ drug candidates. PET is associated with a high level of translatability between species as similar, non-invasive protocols allow for longitudinal rather than cross-sectional studies and can be used both in a preclinical and clinical setting. This review focuses on the use of preclinical PET imaging in genetically modified animals that express human Aβ, and its present and potential future role in the development of drugs aimed at reducing brain Aβ levels as a therapeutic strategy to halt disease progression in AD.
Collapse
Affiliation(s)
- Stina Syvänen
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden.
| | - Silvio R Meier
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Sahar Roshanbin
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Mengfei Xiong
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Rebecca Faresjö
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Tobias Gustavsson
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Gillian Bonvicini
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden.,BioArctic AB, Stockholm, Sweden
| | - Eva Schlein
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Ximena Aguilar
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Ulrika Julku
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Jonas Eriksson
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden.,PET Centre, Uppsala University Hospital, Uppsala, Sweden
| | - Dag Sehlin
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| |
Collapse
|
4
|
Lewis MR, Cutler CS, Jurisson SS. Targeted Antibodies and Peptides. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00022-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
5
|
Krasnovskaya O, Spector D, Zlobin A, Pavlov K, Gorelkin P, Erofeev A, Beloglazkina E, Majouga A. Metals in Imaging of Alzheimer's Disease. Int J Mol Sci 2020; 21:E9190. [PMID: 33276505 PMCID: PMC7730413 DOI: 10.3390/ijms21239190] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 11/25/2020] [Accepted: 11/28/2020] [Indexed: 12/23/2022] Open
Abstract
One of the hallmarks of Alzheimer's disease (AD) is the deposition of amyloid plaques in the brain parenchyma, which occurs 7-15 years before the onset of cognitive symptoms of the pathology. Timely diagnostics of amyloid formations allows identifying AD at an early stage and initiating inhibitor therapy, delaying the progression of the disease. However, clinically used radiopharmaceuticals based on 11C and 18F are synchrotron-dependent and short-lived. The design of new metal-containing radiopharmaceuticals for AD visualization is of interest. The development of coordination compounds capable of effectively crossing the blood-brain barrier (BBB) requires careful selection of a ligand moiety, a metal chelating scaffold, and a metal cation, defining the method of supposed Aβ visualization. In this review, we have summarized metal-containing drugs for positron emission tomography (PET), magnetic resonance imaging (MRI), and single-photon emission computed tomography (SPECT) imaging of Alzheimer's disease. The obtained data allow assessing the structure-ability to cross the BBB ratio.
Collapse
Affiliation(s)
- Olga Krasnovskaya
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory 1,3, 119991 Moscow, Russia; (A.Z.); (K.P.); (P.G.); (A.E.); (E.B.); (A.M.)
- Department of Materials Science of Semiconductors and Dielectrics, National University of Science and Technology (MISIS), Leninskiy Prospect 4, 101000 Moscow, Russia
| | - Daniil Spector
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory 1,3, 119991 Moscow, Russia; (A.Z.); (K.P.); (P.G.); (A.E.); (E.B.); (A.M.)
- Department of Materials Science of Semiconductors and Dielectrics, National University of Science and Technology (MISIS), Leninskiy Prospect 4, 101000 Moscow, Russia
| | - Alexander Zlobin
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory 1,3, 119991 Moscow, Russia; (A.Z.); (K.P.); (P.G.); (A.E.); (E.B.); (A.M.)
| | - Kirill Pavlov
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory 1,3, 119991 Moscow, Russia; (A.Z.); (K.P.); (P.G.); (A.E.); (E.B.); (A.M.)
| | - Peter Gorelkin
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory 1,3, 119991 Moscow, Russia; (A.Z.); (K.P.); (P.G.); (A.E.); (E.B.); (A.M.)
- Department of Materials Science of Semiconductors and Dielectrics, National University of Science and Technology (MISIS), Leninskiy Prospect 4, 101000 Moscow, Russia
| | - Alexander Erofeev
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory 1,3, 119991 Moscow, Russia; (A.Z.); (K.P.); (P.G.); (A.E.); (E.B.); (A.M.)
- Department of Materials Science of Semiconductors and Dielectrics, National University of Science and Technology (MISIS), Leninskiy Prospect 4, 101000 Moscow, Russia
| | - Elena Beloglazkina
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory 1,3, 119991 Moscow, Russia; (A.Z.); (K.P.); (P.G.); (A.E.); (E.B.); (A.M.)
| | - Alexander Majouga
- Chemistry Department, Lomonosov Moscow State University, Leninskie Gory 1,3, 119991 Moscow, Russia; (A.Z.); (K.P.); (P.G.); (A.E.); (E.B.); (A.M.)
- Department of Materials Science of Semiconductors and Dielectrics, National University of Science and Technology (MISIS), Leninskiy Prospect 4, 101000 Moscow, Russia
- Mendeleev University of Chemical Technology of Russia, Miusskaya Ploshchad’ 9, 125047 Moscow, Russia
| |
Collapse
|
6
|
Focused ultrasound for opening blood-brain barrier and drug delivery monitored with positron emission tomography. J Control Release 2020; 324:303-316. [DOI: 10.1016/j.jconrel.2020.05.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/14/2022]
|
7
|
Sedgwick AC, Brewster JT, Harvey P, Iovan DA, Smith G, He XP, Tian H, Sessler JL, James TD. Metal-based imaging agents: progress towards interrogating neurodegenerative disease. Chem Soc Rev 2020; 49:2886-2915. [DOI: 10.1039/c8cs00986d] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Transition metals and lanthanide ions display unique properties that enable the development of non-invasive diagnostic tools for imaging. In this review, we highlight various metal-based imaging strategies used to interrogate neurodegeneration.
Collapse
Affiliation(s)
- Adam C. Sedgwick
- Department of Chemistry
- The University of Texas at Austin
- Austin
- USA
| | | | - Peter Harvey
- Department of Biological Engineering
- Massachusetts Institute of Technology
- Cambridge
- USA
- Sir Peter Mansfield Imaging Centre
| | - Diana A. Iovan
- Department of Chemistry
- University of California
- Berkeley
- USA
| | - Graham Smith
- Division of Radiotherapy & Imaging
- Institute of Cancer Research
- London
- UK
| | - Xiao-Peng He
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering
- Feringa Nobel Prize Scientist Joint Research Center
- School of Chemistry and Molecular Engineering
- East China University of Science and Technology
- Shanghai 200237
| | - He Tian
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering
- Feringa Nobel Prize Scientist Joint Research Center
- School of Chemistry and Molecular Engineering
- East China University of Science and Technology
- Shanghai 200237
| | | | | |
Collapse
|
8
|
Engineered antibodies: new possibilities for brain PET? Eur J Nucl Med Mol Imaging 2019; 46:2848-2858. [PMID: 31342134 PMCID: PMC6879437 DOI: 10.1007/s00259-019-04426-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 07/03/2019] [Indexed: 12/18/2022]
Abstract
Almost 50 million people worldwide are affected by Alzheimer’s disease (AD), the most common neurodegenerative disorder. Development of disease-modifying therapies would benefit from reliable, non-invasive positron emission tomography (PET) biomarkers for early diagnosis, monitoring of disease progression, and assessment of therapeutic effects. Traditionally, PET ligands have been based on small molecules that, with the right properties, can penetrate the blood–brain barrier (BBB) and visualize targets in the brain. Recently a new class of PET ligands based on antibodies have emerged, mainly in applications related to cancer. While antibodies have advantages such as high specificity and affinity, their passage across the BBB is limited. Thus, to be used as brain PET ligands, antibodies need to be modified for active transport into the brain. Here, we review the development of radioligands based on antibodies for visualization of intrabrain targets. We focus on antibodies modified into a bispecific format, with the capacity to undergo transferrin receptor 1 (TfR1)-mediated transcytosis to enter the brain and access pathological proteins, e.g. amyloid-beta. A number of such antibody ligands have been developed, displaying differences in brain uptake, pharmacokinetics, and ability to bind and visualize the target in the brain of transgenic mice. Potential pathological changes related to neurodegeneration, e.g. misfolded proteins and neuroinflammation, are suggested as future targets for this novel type of radioligand. Challenges are also discussed, such as the temporal match of radionuclide half-life with the ligand’s pharmacokinetic profile and translation to human use. In conclusion, brain PET imaging using bispecific antibodies, modified for receptor-mediated transcytosis across the BBB, is a promising method for specifically visualizing molecules in the brain that are difficult to target with traditional small molecule ligands.
Collapse
|
9
|
Jang JH, Han SJ, Kim JY, Kim KI, Lee KC, Kang CS. Synthesis and Feasibility Evaluation of a new Trastuzumab Conjugate Integrated with Paclitaxel and 89Zr for Theranostic Application Against HER2-Expressing Breast Cancers. ChemistryOpen 2019; 8:451-456. [PMID: 31008009 PMCID: PMC6454217 DOI: 10.1002/open.201900037] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/12/2019] [Indexed: 01/26/2023] Open
Abstract
The preparation and in vitro evaluation of a theranostic conjugate composed of trastuzumab, paclitaxel (PTX), and deferoxamine (DFO)-chelated 89Zr have been reported. These comounds have potential applications against HER2 receptor positive breast cancers. We conjugated DFO and PTX to trastuzumab by exploiting simple conjugation chemistry. The conjugate (DFO-trastuzumab-PTX) showed excellent radiolabeling efficiency with 89Zr and the labeled conjugate had high in vitro stability in human serum. Furthermore, DFO-trastuzumab-PTX displayed comparable cytotoxicity with PTX and 89Zr-DFO-trastuzumab-PTX exhibited HER2 receptor-mediated binding on HER2-positive MDA-MB-231 breast cancer cells. The results of our in vitro study indicate high potential of 89Zr-DFO-trastuzumab-PTX to be utilized in the theranostic application against HER2-postive breast cancers.
Collapse
Affiliation(s)
- Joo Hee Jang
- Division of Applied RIKorea Institute of Radiological and Medical Sciences75 Nowon-ro, Nowon-guSeoulKorea01812
| | - Sang Jin Han
- Division of Applied RIKorea Institute of Radiological and Medical Sciences75 Nowon-ro, Nowon-guSeoulKorea01812
| | - Jung Young Kim
- Division of Applied RIKorea Institute of Radiological and Medical Sciences75 Nowon-ro, Nowon-guSeoulKorea01812
| | - Kwang Il Kim
- Division of Applied RIKorea Institute of Radiological and Medical Sciences75 Nowon-ro, Nowon-guSeoulKorea01812
| | - Kyo Chul Lee
- Division of Applied RIKorea Institute of Radiological and Medical Sciences75 Nowon-ro, Nowon-guSeoulKorea01812
| | - Chi Soo Kang
- Division of Applied RIKorea Institute of Radiological and Medical Sciences75 Nowon-ro, Nowon-guSeoulKorea01812
| |
Collapse
|
10
|
Tang Y, Hu Y, Liu W, Chen L, Zhao Y, Ma H, Yang J, Yang Y, Liao J, Cai J, Chen Y, Liu N. A radiopharmaceutical [ 89Zr]Zr-DFO-nimotuzumab for immunoPET with epidermal growth factor receptor expression in vivo. Nucl Med Biol 2019; 70:23-31. [PMID: 30826708 DOI: 10.1016/j.nucmedbio.2019.01.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/04/2019] [Accepted: 01/20/2019] [Indexed: 01/04/2023]
Abstract
INTRODUCTION The potential of the positron-emitting zirconium-89 (89Zr) (t1/2 = 78.4 h) has been recently reported for immune positron emission tomography (immunoPET) radioimmunoconjugates design. In our work, we explored the optimized preparation of [89Zr]Zr-DFO-nimotuzumab, and evaluated 89Zr-labeled monoclonal antibody (mAb) construct for targeted imaging of epidermal growth factor receptor (EGFR) overexpressed in glioma. METHODS To optimize the radiolabeling efficiency of 89Zr with DFO-nimotuzumab, multiple immunoconjugates and radiolabeling were performed. Radiolabeling yield, radiochemical purity, stability, and activity assay were investigated to characterize [89Zr]Zr-DFO-nimotuzumab for chemical and biological integrity. The in vivo behavior of this tracer was studied in mice bearing subcutaneous U87MG (EGFR-positive) tumors received a 3.5 ± 0.2 MBq/dose using PET/CT imaging. One group mice bearing subcutaneous U87MG (EGFR-positive) tumors received [89Zr]Zr-DFO-nimotuzumab (3.5 ± 0.2 MBq, ~3 μg) (nonblocking) for immunoPET; the other group had 30 μg predose (blocking) of cold nimotuzumab 24 h prior to [89Zr]Zr-DFO-nimotuzumab. RESULTS [89Zr]Zr-DFO-nimotuzumab was prepared with high radiochemical yield (>90%), radiochemical purity (>99%), and specific activity (115 ± 0.8 MBq/mg). In vitro validation showed that [89Zr]Zr-DFO-nimotuzumab had an initial immunoreactive fraction of 0.99 ± 0.05 and remained active for up to 5 days. A biodistribution study revealed excellent stability of [89Zr]Zr-DFO-nimotuzumab in vivo compared with 89Zr as a bone seeker. High uptake in the liver and heart and modest penetration in the brain were observed, with no significant accumulation of activity in other organs. ImmunoPET studies also indicated prominent image contrast that remarkably high uptake up to ~20%ID/g for nonblocking and ~2%ID/g for blocking in tumor between 12 and 120 h after administration. CONCLUSION These studies developed a radiopharmaceutical [89Zr]Zr-DFO-nimotuzumab with optimized synthesis. The potential utility of [89Zr]Zr-DFO-nimotuzumab in assessing EGFR status in glioma was demonstrated in this study.
Collapse
Affiliation(s)
- Yu Tang
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, PR China; Department of Nuclear Medicine, Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou 646000, PR China; Chengdu New Radiomedicine Technology Co. Ltd., Chengdu 610000, PR China
| | - Yingjiang Hu
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, PR China
| | - Weihao Liu
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, PR China
| | - Lin Chen
- Department of Nuclear Medicine, Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou 646000, PR China
| | - Yan Zhao
- Department of Nuclear Medicine, Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou 646000, PR China
| | - Huan Ma
- Chengdu New Radiomedicine Technology Co. Ltd., Chengdu 610000, PR China
| | - Jijun Yang
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, PR China
| | - Yuanyou Yang
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, PR China.
| | - Jiali Liao
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, PR China
| | - Jiming Cai
- Chengdu New Radiomedicine Technology Co. Ltd., Chengdu 610000, PR China
| | - Yue Chen
- Department of Nuclear Medicine, Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou 646000, PR China
| | - Ning Liu
- Key Laboratory of Radiation Physics and Technology (Sichuan University), Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, PR China.
| |
Collapse
|
11
|
De Vijlder T, Fissers J, Van Broeck B, Wyffels L, Mercken M, Pemberton DJ. Mass spectrometric characterization of intact desferal-conjugated monoclonal antibodies for immuno-PET imaging. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2018; 32:1643-1650. [PMID: 29943865 DOI: 10.1002/rcm.8209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 06/03/2018] [Accepted: 06/12/2018] [Indexed: 06/08/2023]
Abstract
RATIONALE Immuno-PET imaging may prove to be a diagnostic and progression/intervention biomarker for Alzheimer's disease (AD) with improved sensitivity and specificity. Immuno-PET imaging is based on the coupling of an antibody with a chelator that captures a radioisotope thus serving as an in-vivo PET ligand. A robust and quality controlled process for linking the chelator to the-antibody is fundamental for the success of this approach. METHODS The structural integrities of two monoclonal antibodies (trastuzumab and JRF/AβN/25) and the quantity of desferal-based chelator attached following modification of the antibodies were assessed by online desalting and intact mass analysis. Enzymatic steps for the deglycosylation and removal of C-terminal lysine was performed sequentially and in a single tube to improve intact mass data. RESULTS Intact mass analysis demonstrated that inclusion of enzymatic processing was critical to correctly derive the quantity of chelator linked to the monoclonal antibodies. For trastuzumab, enzymatic cleaving of the glycans was sufficient, whilst additional removal of the C-terminal lysine was necessary for JRF/AβN/25 to ensure reproducible assessment of the relatively low amount of attached chelator. CONCLUSIONS An efficient intact mass analysis-based process was developed to reproducibly determine the integrity of monoclonal antibodies and the quantity of attached chelator. This technique could serve as an essential quality control approach for the development and production of immuno-PET tracers.
Collapse
Affiliation(s)
- Thomas De Vijlder
- Pharmaceutical Development & Manufacturing Sciences, Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Jens Fissers
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Bianca Van Broeck
- Neuroscience Discovery, Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Leonie Wyffels
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
- Department of Nuclear Medicine, University Hospital Antwerp, Edegem, Belgium
| | - Marc Mercken
- Neuroscience Discovery, Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Darrel J Pemberton
- Neuroscience Experimental Medicine, Janssen Pharmaceutica NV, Beerse, Belgium
| |
Collapse
|
12
|
Chauhan K, Tiwari AK, Chadha N, Kaul A, Singh AK, Datta A. Chalcone Based Homodimeric PET Agent, 11C-(Chal) 2DEA-Me, for Beta Amyloid Imaging: Synthesis and Bioevaluation. Mol Pharm 2018. [PMID: 29522675 DOI: 10.1021/acs.molpharmaceut.7b01070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Homodimeric chalcone based 11C-PET radiotracer, 11C-(Chal)2DEA-Me, was synthesized, and binding affinity toward beta amyloid (Aβ) was evaluated. The computational studies revealed multiple binding of the tracer at the recognition sites of Aβ fibrils. The bivalent ligand 11C-(Chal)2DEA-Me displayed higher binding affinity compared to the corresponding monomer, 11C-Chal-Me, and classical Aβ agents. The radiolabeling yield with carbon-11 was 40-55% (decay corrected) with specific activity of 65-90 GBq/μmol. A significant ( p < 0.0001) improvement in the binding affinity of 11C-(Chal)2DEA-Me with synthetic Aβ42 aggregates over the monomer, 11C-Chal-Me, demonstrates the utility of the bivalent approach. The PET imaging and biodistribution data displayed suitable brain pharmacokinetics of both ligands with higher brain uptake in the case of the bivalent ligand. Metabolite analysis of healthy ddY mouse brain homogenates exhibited high stability of the radiotracers in the brain with >93% intact tracer at 30 min post injection. Both chalcone derivatives were fluorescent in nature and demonstrated significant changes in the emission properties after binding with Aβ42. The preliminary analysis indicates high potential of 11C-(Chal)2DEA-Me as in vivo Aβ42 imaging tracer and highlights the significance of the bivalent approach to achieve a higher biological response for detection of early stages of amyloidosis.
Collapse
Affiliation(s)
- Kanchan Chauhan
- Division of Cyclotron and Radiopharmaceutical Sciences , Institute of Nuclear Medicine & Allied Sciences, DRDO , Brig. SK Mazumdar Marg , Delhi 110054 , India.,Departamento de Bionanotecnología, Centro de Nanociencias y Nanotecnología , Universidad Nacional Autónoma de México , Km. 107 Carratera Tijuana-Ensenada , 22860 Ensenada , Baja California , Mexico
| | - Anjani K Tiwari
- Division of Cyclotron and Radiopharmaceutical Sciences , Institute of Nuclear Medicine & Allied Sciences, DRDO , Brig. SK Mazumdar Marg , Delhi 110054 , India.,Department of Applied Chemistry , Babasaheb Bhimrao Ambedkar University , Lucknow , India
| | - Nidhi Chadha
- Division of Cyclotron and Radiopharmaceutical Sciences , Institute of Nuclear Medicine & Allied Sciences, DRDO , Brig. SK Mazumdar Marg , Delhi 110054 , India
| | - Ankur Kaul
- Division of Cyclotron and Radiopharmaceutical Sciences , Institute of Nuclear Medicine & Allied Sciences, DRDO , Brig. SK Mazumdar Marg , Delhi 110054 , India
| | - Ajai Kumar Singh
- Department of Chemistry , Indian Institute of Technology , Delhi 110016 , India
| | - Anupama Datta
- Division of Cyclotron and Radiopharmaceutical Sciences , Institute of Nuclear Medicine & Allied Sciences, DRDO , Brig. SK Mazumdar Marg , Delhi 110054 , India
| |
Collapse
|
13
|
Amyloid β-targeted metal complexes for potential applications in Alzheimer's disease. Future Med Chem 2018; 10:679-701. [DOI: 10.4155/fmc-2017-0248] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is currently an incurable neurodegenerative disorder that affects millions of people around the world. The aggregation of amyloid-β peptides (Aβ), one of the primary pathological hallmarks of AD, plays a key role in the AD pathogenesis. In this regard, Aβ aggregates have been considered as both biomarkers and drug targets for the diagnosis and therapy of AD. Various Aβ-targeted metal complexes have exhibited promising potential as anti-AD agents due to their fascinating physicochemical properties over the past two decades. This review classifies the complexes into three groups based on their potential applications in AD including therapy, diagnosis and theranosis. The recent representative examples are highlighted in terms of design rationale, working mechanism and potential applications.
Collapse
|
14
|
Jalilian AR, Osso JA. Production, applications and status of zirconium-89 immunoPET agents. J Radioanal Nucl Chem 2017. [DOI: 10.1007/s10967-017-5358-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
15
|
Chen K, Cui M. Recent progress in the development of metal complexes as β-amyloid imaging probes in the brain. MEDCHEMCOMM 2017; 8:1393-1407. [PMID: 30108850 PMCID: PMC6072098 DOI: 10.1039/c7md00064b] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 05/11/2017] [Indexed: 01/28/2023]
Abstract
In this review, we have focused on the recent progress in metal complexes that are able to bind to β-amyloid (Aβ) species. We have discussed various radioactive complexes of 99mTc, 68Ga, 64Cu, 89Zr, and 111In, which were designed as Aβ imaging agents for positron emission tomography (PET) and single photon emission computed tomography (SPECT) imaging, non-radioactive Re and Ru complexes as Aβ sensors using luminescence methods, and Gd3+ complexes as contrast agents for magnetic resonance imaging (MRI).
Collapse
Affiliation(s)
- Kaihua Chen
- Key Laboratory of Radiopharmaceuticals , Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , P. R. China . ; ; Tel: +86 10 58808891
| | - Mengchao Cui
- Key Laboratory of Radiopharmaceuticals , Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , P. R. China . ; ; Tel: +86 10 58808891
| |
Collapse
|
16
|
Lacerda S, Morfin JF, Geraldes CFGC, Tóth É. Metal complexes for multimodal imaging of misfolded protein-related diseases. Dalton Trans 2017; 46:14461-14474. [DOI: 10.1039/c7dt02371e] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aggregation of misfolded proteins and progressive polymerization of otherwise soluble proteins is a common hallmark of several highly debilitating and increasingly prevalent diseases, including amyotrophic lateral sclerosis, cerebral amyloid angiopathy, type II diabetes and Parkinson's, Huntington's and Alzheimer's diseases.
Collapse
Affiliation(s)
- S. Lacerda
- Centre de Biophysique Moléculaire
- CNRS
- UPR 4301
- Université d'Orléans
- 45071 Orléans Cedex 2
| | - J.-F. Morfin
- Centre de Biophysique Moléculaire
- CNRS
- UPR 4301
- Université d'Orléans
- 45071 Orléans Cedex 2
| | - C. F. G. C. Geraldes
- Department of Life Sciences
- Faculty of Sciences and Technology
- University of Coimbra
- 3000-393 Coimbra
- Portugal
| | - É. Tóth
- Centre de Biophysique Moléculaire
- CNRS
- UPR 4301
- Université d'Orléans
- 45071 Orléans Cedex 2
| |
Collapse
|
17
|
wyffels L, Verbrugghen T, Monnery BD, Glassner M, Stroobants S, Hoogenboom R, Staelens S. μPET imaging of the pharmacokinetic behavior of medium and high molar mass 89 Zr-labeled poly(2-ethyl-2-oxazoline) in comparison to poly(ethylene glycol). J Control Release 2016; 235:63-71. [DOI: 10.1016/j.jconrel.2016.05.048] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/20/2016] [Accepted: 05/21/2016] [Indexed: 01/20/2023]
|
18
|
Waldron AM, Fissers J, Van Eetveldt A, Van Broeck B, Mercken M, Pemberton DJ, Van Der Veken P, Augustyns K, Joossens J, Stroobants S, Dedeurwaerdere S, Wyffels L, Staelens S. In Vivo Amyloid-β Imaging in the APPPS1-21 Transgenic Mouse Model with a (89)Zr-Labeled Monoclonal Antibody. Front Aging Neurosci 2016; 8:67. [PMID: 27064204 PMCID: PMC4815004 DOI: 10.3389/fnagi.2016.00067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 03/21/2016] [Indexed: 11/30/2022] Open
Abstract
Introduction: The accumulation of amyloid-β is a pathological hallmark of Alzheimer’s disease and is a target for molecular imaging probes to aid in diagnosis and disease monitoring. This study evaluated the feasibility of using a radiolabeled monoclonal anti-amyloid-β antibody (JRF/AβN/25) to non-invasively assess amyloid-β burden in aged transgenic mice (APPPS1–21) with μPET imaging. Methods: We investigated the antibody JRF/AβN/25 that binds to full-length Aβ. JRF/AβN/25 was radiolabeled with a [89Zr]-desferal chelate and intravenously injected into 12–13 month aged APPPS1–21 mice and their wild-type (WT) controls. Mice underwent in vivo μPET imaging at 2, 4, and 7 days post injection and were sacrificed at the end of each time point to assess brain penetrance, plaque labeling, biodistribution, and tracer stability. To confirm imaging specificity we also evaluated brain uptake of a non-amyloid targeting [89Zr]-labeled antibody (trastuzumab) as a negative control, additionally we performed a competitive blocking study with non-radiolabeled Df-Bz-JRF/AβN/25 and finally we assessed the possible confounding effects of blood retention. Results: Voxel-wise analysis of μPET data demonstrated significant [89Zr]-Df-Bz-JRF/AβN/25 retention in APPPS1–21 mice at all time points investigated. With ex vivo measures of radioactivity, significantly higher retention of [89Zr]-Df-Bz-JRF/AβN/25 was found at 4 and 7 days pi in APPPS1–21 mice. Despite the observed genotypic differences, comparisons with immunohistochemistry revealed that in vivo plaque labeling was low. Furthermore, pre-treatment with Df-Bz-JRF/AβN/25 only partially blocked [89Zr]-Df-Bz-JRF/AβN/25 uptake indicative of a high contribution of non-specific binding. Conclusion: Amyloid plaques were detected in vivo with a radiolabeled monoclonal anti-amyloid antibody. The low brain penetrance of the antibody in addition to non-specific binding prevented an accurate estimation of plaque burden. However, it should be noted that [89Zr]-Df-Bz-JRF/AβN/25 nevertheless demonstrated in vivo binding and strategies to increase brain penetrance would likely achieve better results.
Collapse
Affiliation(s)
- Ann-Marie Waldron
- Faculty of Medicine and Health Sciences, Molecular Imaging Center Antwerp, University of AntwerpAntwerp, Belgium; Translational Neurosciences, University of AntwerpAntwerp, Belgium
| | - Jens Fissers
- Faculty of Medicine and Health Sciences, Molecular Imaging Center Antwerp, University of AntwerpAntwerp, Belgium; Laboratory of Medicinal Chemistry, University of AntwerpAntwerp, Belgium
| | | | - Bianca Van Broeck
- Department of Neuroscience, Janssen Research and Development, A Division of Janssen Pharmaceutica NV Beerse, Belgium
| | - Marc Mercken
- Department of Neuroscience, Janssen Research and Development, A Division of Janssen Pharmaceutica NV Beerse, Belgium
| | - Darrel J Pemberton
- Department of Neuroscience, Janssen Research and Development, A Division of Janssen Pharmaceutica NV Beerse, Belgium
| | | | - Koen Augustyns
- Laboratory of Medicinal Chemistry, University of Antwerp Antwerp, Belgium
| | - Jurgen Joossens
- Laboratory of Medicinal Chemistry, University of Antwerp Antwerp, Belgium
| | | | | | - Leonie Wyffels
- Faculty of Medicine and Health Sciences, Molecular Imaging Center Antwerp, University of AntwerpAntwerp, Belgium; Nuclear Medicine, University Hospital AntwerpAntwerp, Belgium
| | - Steven Staelens
- Faculty of Medicine and Health Sciences, Molecular Imaging Center Antwerp, University of Antwerp Antwerp, Belgium
| |
Collapse
|